51
|
Xie K, Wang Z, Qi L, Zhao X, Wang Y, Qu J, Xu P, Huang L, Zhang W, Yang Y, Wang X, Shi P. Profiling MicroRNAs with Associated Spatial Dynamics in Acute Tissue Slices. ACS NANO 2021; 15:4881-4892. [PMID: 33719400 DOI: 10.1021/acsnano.0c09676] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
MicroRNAs (miRNAs) are suggested to play important roles in the pathogenesis and progress of human diseases with heterogeneous regulation in different types of cells. However, limited technique is available for profiling miRNAs with both expression and spatial dynamics. Here, we describe a platform for multiplexed in situ miRNA profiling in acute tissue slices. The technique uses diamond nanoneedles functionalized with RNA-binding proteins to directly isolate targeted miRNAs from the cytosol of a large population of cells to achieve a quasi-single-cell analysis for a tissue sample. In addition to a quantitative evaluation of the expression level of particular miRNAs, the technique also provides the relative spatial dynamics of the cellular miRNAs in associated cell populations, which was demonstrated to be useful in analyzing the susceptibility and spatial reorganization of different types of cells in the tissues from normal or diseased animals. As a proof-of-concept, in MK-801-induced schizophrenia model, we found that astrocytes, instead of neurons, are more heterogeneously affected in the hippocampus of rats that underwent repeated injection of MK-801, showing an expression fingerprint related to differentially down-regulated miRNA-135a and miRNA-143; the associated astrocyte subpopulation is also more spatially dispersed in the hippocampus, suggesting an astrocyte dysregulation in the induced schizophrenia animals.
Collapse
Affiliation(s)
- Kai Xie
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR China
| | - Zixun Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR China
| | - Lin Qi
- Department of Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong SAR China
| | - Xi Zhao
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR China
| | - Yuan Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR China
| | - Jin Qu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR China
| | - Ping Xu
- Department of Respiratory and Critical Care Medicine, Peking University Shenzhen Hospital, Shenzhen, China 518036
| | - Linfeng Huang
- Division of Natural and Applied Sciences, Duke Kunshan University, Kunshan, Jiangsu China 215300
| | - Wenjun Zhang
- Department of Materials Science and Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR China
- Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, Kowloon, Hong Kong SAR China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China 518000
| | - Yang Yang
- Functional Thin Films Research Center, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China 518055
- Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, Kowloon, Hong Kong SAR China
| | - Xin Wang
- Department of Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong SAR China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China 518000
| | - Peng Shi
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR China
- Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, Kowloon, Hong Kong SAR China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China 518000
| |
Collapse
|
52
|
Yue W, Sun J, Zhang J, Chang Y, Shen Q, Zhu Z, Yu S, Wu X, Peng S, Li N, Hua J. Mir-34c affects the proliferation and pluripotency of porcine induced pluripotent stem cell (piPSC)-like cells by targeting c-Myc. Cells Dev 2021; 166:203665. [PMID: 33994350 DOI: 10.1016/j.cdev.2021.203665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/30/2021] [Accepted: 02/09/2021] [Indexed: 12/28/2022]
Abstract
MicroRNAs are important regulators in stem cells, which involve in gene regulation, including cell proliferation, differentiation and apoptosis. As an important one, miR-34c participates in various processes by targeting protein-coding genes. It is generally considered as a tumor suppressor and cell adhesion inhibitor. However, whether miR-34c has effects on pluripotent stem cells is not clear. Here, by mir-34c mimics transfection, the function of miR-34c on porcine induced pluripotent stem cell (piPSC)-like cells was investigated. Bioinformatics analyses showed that c-Myc is miR-34c's candidate target, which was confirmed by dual Luciferase assay. The knockout of miR-34c indicated that mir-34c affects the proliferation and pluripotency of piPSC-like cells by targeting c-Myc. Our study explored the regulatory mechanism of miR-34c on piPSC-like cells, providing a reference for the establishment of true porcine PSCs.
Collapse
Affiliation(s)
- Wei Yue
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jing Sun
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Juqing Zhang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yongxing Chang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Qiaoyan Shen
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zhenshuo Zhu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Shuai Yu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiaolong Wu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Sha Peng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Na Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
53
|
Segaran RC, Chan LY, Wang H, Sethi G, Tang FR. Neuronal Development-Related miRNAs as Biomarkers for Alzheimer's Disease, Depression, Schizophrenia and Ionizing Radiation Exposure. Curr Med Chem 2021; 28:19-52. [PMID: 31965936 DOI: 10.2174/0929867327666200121122910] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 09/30/2019] [Accepted: 10/22/2019] [Indexed: 11/22/2022]
Abstract
Radiation exposure may induce Alzheimer's disease (AD), depression or schizophrenia. A number of experimental and clinical studies suggest the involvement of miRNA in the development of these diseases, and also in the neuropathological changes after brain radiation exposure. The current literature review indicated the involvement of 65 miRNAs in neuronal development in the brain. In the brain tissue, blood, or cerebral spinal fluid (CSF), 11, 55, or 28 miRNAs are involved in the development of AD respectively, 89, 50, 19 miRNAs in depression, and 102, 35, 8 miRNAs in schizophrenia. We compared miRNAs regulating neuronal development to those involved in the genesis of AD, depression and schizophrenia and also those driving radiation-induced brain neuropathological changes by reviewing the available data. We found that 3, 11, or 8 neuronal developmentrelated miRNAs from the brain tissue, 13, 16 or 14 miRNAs from the blood of patient with AD, depression and schizophrenia respectively were also involved in radiation-induced brain pathological changes, suggesting a possibly specific involvement of these miRNAs in radiation-induced development of AD, depression and schizophrenia respectively. On the other hand, we noted that radiationinduced changes of two miRNAs, i.e., miR-132, miR-29 in the brain tissue, three miRNAs, i.e., miR- 29c-5p, miR-106b-5p, miR-34a-5p in the blood were also involved in the development of AD, depression and schizophrenia, thereby suggesting that these miRNAs may be involved in the common brain neuropathological changes, such as impairment of neurogenesis and reduced learning memory ability observed in these three diseases and also after radiation exposure.
Collapse
Affiliation(s)
- Renu Chandra Segaran
- Radiation Physiology Lab, Singapore Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, Singapore 138602, Singapore
| | - Li Yun Chan
- Radiation Physiology Lab, Singapore Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, Singapore 138602, Singapore
| | - Hong Wang
- Radiation Physiology Lab, Singapore Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, Singapore 138602, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Feng Ru Tang
- Radiation Physiology Lab, Singapore Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, Singapore 138602, Singapore
| |
Collapse
|
54
|
MicroRNAs as Candidate Biomarkers for Alzheimer's Disease. Noncoding RNA 2021; 7:ncrna7010008. [PMID: 33535543 PMCID: PMC7930943 DOI: 10.3390/ncrna7010008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/14/2021] [Accepted: 01/28/2021] [Indexed: 12/17/2022] Open
Abstract
The neurological damage of Alzheimer’s disease (AD) is thought to be irreversible upon onset of dementia-like symptoms, as it takes years to decades for occult pathologic changes to become symptomatic. It is thus necessary to identify individuals at risk for the development of the disease before symptoms manifest in order to provide early intervention. Surrogate markers are critical for early disease detection, stratification of patients in clinical trials, prediction of disease progression, evaluation of response to treatment, and also insight into pathomechanisms. Here, we review the evidence for a number of microRNAs that may serve as biomarkers with possible mechanistic insights into the AD pathophysiologic processes, years before the clinical manifestation of the disease.
Collapse
|
55
|
Gentile G, Paciello F, Zorzi V, Spampinato AG, Guarnaccia M, Crispino G, Tettey-Matey A, Scavizzi F, Raspa M, Fetoni AR, Cavallaro S, Mammano F. miRNA and mRNA Profiling Links Connexin Deficiency to Deafness via Early Oxidative Damage in the Mouse Stria Vascularis. Front Cell Dev Biol 2021; 8:616878. [PMID: 33569381 PMCID: PMC7868390 DOI: 10.3389/fcell.2020.616878] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/10/2020] [Indexed: 12/15/2022] Open
Abstract
Pathogenic mutations in the non-syndromic hearing loss and deafness 1 (DFNB1) locus are the primary cause of monogenic inheritance for prelingual hearing loss. To unravel molecular pathways involved in etiopathology and look for early degeneration biomarkers, we used a system biology approach to analyze Cx30−/− mice at an early cochlear post-natal developmental stage. These mice are a DFNB1 mouse model with severely reduced expression levels of two connexins in the inner ear, Cx30, and Cx26. Integrated analysis of miRNA and mRNA expression profiles in the cochleae of Cx30−/− mice at post-natal day 5 revealed the overexpression of five miRNAs (miR-34c, miR-29b, miR-29c, miR-141, and miR-181a) linked to apoptosis, oxidative stress, and cochlear degeneration, which have Sirt1 as a common target of transcriptional and/or post-transcriptional regulation. In young adult Cx30−/− mice (3 months of age), these alterations culminated with blood barrier disruption in the Stria vascularis (SV), which is known to have the highest aerobic metabolic rate of all cochlear structures and whose microvascular alterations contribute to age-related degeneration and progressive decline of auditory function. Our experimental validation of selected targets links hearing acquisition failure in Cx30−/− mice, early oxidative stress, and metabolic dysregulation to the activation of the Sirt1–p53 axis. This is the first integrated analysis of miRNA and mRNA in the cochlea of the Cx30−/− mouse model, providing evidence that connexin downregulation determines a miRNA-mediated response which leads to chronic exhaustion of cochlear antioxidant defense mechanisms and consequent SV dysfunction. Our analyses support the notion that connexin dysfunction intervenes early on during development, causing vascular damage later on in life. This study identifies also early miRNA-mediated biomarkers of hearing impairment, either inherited or age related.
Collapse
Affiliation(s)
- Giulia Gentile
- Department of Biomedical Sciences, National Research Council (CNR) Institute for Biomedical Research and Innovation, Catania, Italy
| | - Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Veronica Zorzi
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy.,Department of Biomedical Sciences, National Research Council (CNR) Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Antonio Gianmaria Spampinato
- Department of Biomedical Sciences, National Research Council (CNR) Institute for Biomedical Research and Innovation, Catania, Italy.,Department of Mathematics and Computer Science, University of Catania, Catania, Italy
| | - Maria Guarnaccia
- Department of Biomedical Sciences, National Research Council (CNR) Institute for Biomedical Research and Innovation, Catania, Italy
| | - Giulia Crispino
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Abraham Tettey-Matey
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Ferdinando Scavizzi
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Marcello Raspa
- Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Anna Rita Fetoni
- Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy.,Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sebastiano Cavallaro
- Department of Biomedical Sciences, National Research Council (CNR) Institute for Biomedical Research and Innovation, Catania, Italy
| | - Fabio Mammano
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Biochemistry and Cell Biology, Rome, Italy.,Department of Physics and Astronomy "G. Galilei", University of Padua, Padua, Italy
| |
Collapse
|
56
|
Santos-Bezerra DP, Cavaleiro AM, Santos AS, Suemoto CK, Pasqualucci CA, Jacob-Filho W, Leite REP, Passarelli M, Marie SKN, Machado UF, Correa-Giannella ML. Alcohol Use Disorder is Associated with Upregulation of MicroRNA-34a and MicroRNA-34c in Hippocampal Postmortem Tissue. Alcohol Clin Exp Res 2021; 45:64-68. [PMID: 33190281 DOI: 10.1111/acer.14505] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/04/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND To investigate epigenetic mechanisms potentially involved in the cognitive decline associated with chronic alcohol intake, we evaluated the expressions of three micro-RNAs (miR-34a, -34b, and -34c) highly expressed in the hippocampus and involved in neuronal physiology and pathology. MiR-34a participates in functioning and survival of mature neurons; miR-34b is associated with Alzheimer-like disorders; and miR-34c is implicated in the memory impairment of Alzheimer disease in rodents and humans. METHODS A total of 69 cases were selected from the Biobank for Aging Studies and categorized according to the absence (n = 50) or presence (n = 19) of alcohol use disorder (AUD). Cases presenting with neuropathological diagnoses of dementias were excluded. Total RNA was extracted from hippocampal paraffinized slices, complementary DNA was synthesized from miRs, and RT-qPCR was performed with TaqMan® assays. RESULTS Higher expressions of miR-34a and miR-34c, but not of miR-34b, were found in the group with AUD in comparison with the group without AUD after adjustment for potential confounders (age, sex, body mass index, presence of hypertension, diabetes mellitus, smoking, and physical inactivity). CONCLUSIONS Hippocampal upregulation of miR-34a and miR-34c may be involved in the cognitive decline associated with chronic alcohol consumption.
Collapse
Affiliation(s)
- Daniele P Santos-Bezerra
- From the Laboratorio de Carboidratos e Radioimunoensaio (LIM-18) do Hospital das Clinicas HCFMUSP, DPS-B, AMC, ASS, MLC-G Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Ana Mercedes Cavaleiro
- From the Laboratorio de Carboidratos e Radioimunoensaio (LIM-18) do Hospital das Clinicas HCFMUSP, DPS-B, AMC, ASS, MLC-G Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Aritania Sousa Santos
- From the Laboratorio de Carboidratos e Radioimunoensaio (LIM-18) do Hospital das Clinicas HCFMUSP, DPS-B, AMC, ASS, MLC-G Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Claudia Kimie Suemoto
- Division of Geriatrics (CKS, WJ-F, REPL), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | | | - Wilson Jacob-Filho
- Division of Geriatrics (CKS, WJ-F, REPL), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Renata Elaine Paraizo Leite
- Division of Geriatrics (CKS, WJ-F, REPL), University of Sao Paulo Medical School, Sao Paulo, Brazil.,Department of Pathology (CAP, REPL), University of Sao Paulo, Sao Paulo, Brazil
| | - Marisa Passarelli
- Laboratorio de Lipides (LIM-10) do HCFMUSP (MP), Faculdade de Medicina, Universidade de São Paulo, Sao Paulo, Brazil.,Programa de Pos-Graduacao em Medicina (MP, MLC-G), Universidade Nove de Julho (UNINOVE), Sao Paulo, Brazil
| | | | - Ubiratan Fabres Machado
- Departmento de Fisiologia e Biofisica (UFM), Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Maria Lucia Correa-Giannella
- From the Laboratorio de Carboidratos e Radioimunoensaio (LIM-18) do Hospital das Clinicas HCFMUSP, DPS-B, AMC, ASS, MLC-G Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil.,Programa de Pos-Graduacao em Medicina (MP, MLC-G), Universidade Nove de Julho (UNINOVE), Sao Paulo, Brazil
| |
Collapse
|
57
|
Kshirsagar V, Thingore C, Juvekar A. Insulin resistance: a connecting link between Alzheimer's disease and metabolic disorder. Metab Brain Dis 2021; 36:67-83. [PMID: 32986168 DOI: 10.1007/s11011-020-00622-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022]
Abstract
Recent evidence suggests that Alzheimer's disease (AD) is closely linked with insulin resistance, as seen in type 2 diabetes mellitus (T2DM). Insulin signaling is impaired in AD brains due to insulin resistance, ultimately resulting in the formation of neurofibrillary tangles (NFTs). AD and T2DM are connected at molecular, clinical, and epidemiological levels making it imperative to understand the contribution of T2DM, and other metabolic disorders, to AD pathogenesis. In this review, we have discussed various modalities involved in the pathogenesis of these two diseases and explained the contributing parameters. Insulin is vital for maintaining glucose homeostasis and it plays an important role in regulating inflammation. Here, we have discussed the roles of various contributing factors like miRNA, leptin hormone, neuroinflammation, metabolic dysfunction, and gangliosides in insulin impairment both in AD and T2DM. Understanding these mechanisms will be a big step forward for making molecular therapies that may help maintain or prevent both AD and T2DM, thus reducing the burden of both these diseases.
Collapse
Affiliation(s)
- Viplav Kshirsagar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Near Khalsa college, Matunga, Mumbai, Maharashtra, 400019, India
| | - Chetan Thingore
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Near Khalsa college, Matunga, Mumbai, Maharashtra, 400019, India
| | - Archana Juvekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Near Khalsa college, Matunga, Mumbai, Maharashtra, 400019, India.
| |
Collapse
|
58
|
MicroRNAs Regulating Autophagy in Neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1208:191-264. [PMID: 34260028 DOI: 10.1007/978-981-16-2830-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Social and economic impacts of neurodegenerative diseases (NDs) become more prominent in our constantly aging population. Currently, due to the lack of knowledge about the aetiology of most NDs, only symptomatic treatment is available for patients. Hence, researchers and clinicians are in need of solid studies on pathological mechanisms of NDs. Autophagy promotes degradation of pathogenic proteins in NDs, while microRNAs post-transcriptionally regulate multiple signalling networks including autophagy. This chapter will critically discuss current research advancements in the area of microRNAs regulating autophagy in NDs. Moreover, we will introduce basic strategies and techniques used in microRNA research. Delineation of the mechanisms contributing to NDs will result in development of better approaches for their early diagnosis and effective treatment.
Collapse
|
59
|
Grinkevich LN. The role of microRNAs in learning and long-term memory. Vavilovskii Zhurnal Genet Selektsii 2020; 24:885-896. [PMID: 35088002 PMCID: PMC8763713 DOI: 10.18699/vj20.687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/11/2020] [Accepted: 10/15/2020] [Indexed: 01/10/2023] Open
Abstract
The mechanisms of long-term memory formation and ways to improve it (in the case of its impairment) remain an extremely difficult problem yet to be solved. Over the recent years, much attention has been
paid to microRNAs in this regard. MicroRNAs are unique endogenous non-coding RNAs about 22 nucleotides in
length; each can regulate translation of hundreds of messenger RNA targets, thereby controlling entire gene networks. MicroRNAs are widely represented in the central nervous system. A large number of studies are currently
being conducted to investigate the role of microRNAs in the brain functioning. A number of microRNAs have
been shown to be involved in the process of synaptic plasticity, as well as in the long-term memory formation.
Disruption of microRNA biogenesis leads to significant cognitive dysfunctions. Moreover, impaired microRNA
biogenesis is one of the causes of the pathogenesis of mental disorders, neurodegenerative illnesses and senile
dementia, which are often accompanied by deterioration in the learning ability and by memory impairment.
Optimistic predictions are made that microRNAs can be used as targets for therapeutic treatment and for diagnosing the above pathologies. The importance of applications related to microRNAs significantly raises interest
in studying their functions in the brain. Thus, this review is focused on the role of microRNAs in cognitive processes. It describes microRNA biogenesis and the role of miRNAs in the regulation of gene expression, as well
as the latest achievements in studying the functional role of microRNAs in learning and in long-term memory
formation, depending on the activation or inhibition of their expression. The review presents summarized data
on the effect of impaired microRNA biogenesis on long-term memory formation, including those associated with
sleep deprivation. In addition, analysis is provided of the current literature related to the prospects of improving
cognitive processes by influencing microRNA biogenesis via the use of CRISPR/Cas9 technologies and active
mental and physical exercises.
Collapse
Affiliation(s)
- L. N. Grinkevich
- Pavlov Institute of Physiology of the Russian Academy of Sciences
| |
Collapse
|
60
|
Culibrk RA, Hahn MS. The Role of Chronic Inflammatory Bone and Joint Disorders in the Pathogenesis and Progression of Alzheimer's Disease. Front Aging Neurosci 2020; 12:583884. [PMID: 33364931 PMCID: PMC7750365 DOI: 10.3389/fnagi.2020.583884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Late-onset Alzheimer's Disease (LOAD) is a devastating neurodegenerative disorder that causes significant cognitive debilitation in tens of millions of patients worldwide. Throughout disease progression, abnormal secretase activity results in the aberrant cleavage and subsequent aggregation of neurotoxic Aβ plaques in the cerebral extracellular space and hyperphosphorylation and destabilization of structural tau proteins surrounding neuronal microtubules. Both pathologies ultimately incite the propagation of a disease-associated subset of microglia-the principle immune cells of the brain-characterized by preferentially pro-inflammatory cytokine secretion and inhibited AD substrate uptake capacity, which further contribute to neuronal degeneration. For decades, chronic neuroinflammation has been identified as one of the cardinal pathophysiological driving features of AD; however, despite a number of works postulating the underlying mechanisms of inflammation-mediated neurodegeneration, its pathogenesis and relation to the inception of cognitive impairment remain obscure. Moreover, the limited clinical success of treatments targeting specific pathological features in the central nervous system (CNS) illustrates the need to investigate alternative, more holistic approaches for ameliorating AD outcomes. Accumulating evidence suggests significant interplay between peripheral immune activity and blood-brain barrier permeability, microglial activation and proliferation, and AD-related cognitive decline. In this work, we review a narrow but significant subset of chronic peripheral inflammatory conditions, describe how these pathologies are associated with the preponderance of neuroinflammation, and posit that we may exploit peripheral immune processes to design interventional, preventative therapies for LOAD. We then provide a comprehensive overview of notable treatment paradigms that have demonstrated considerable merit toward treating these disorders.
Collapse
Affiliation(s)
| | - Mariah S. Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| |
Collapse
|
61
|
Ijomone OM, Ijomone OK, Iroegbu JD, Ifenatuoha CW, Olung NF, Aschner M. Epigenetic influence of environmentally neurotoxic metals. Neurotoxicology 2020; 81:51-65. [PMID: 32882300 PMCID: PMC7708394 DOI: 10.1016/j.neuro.2020.08.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
Continuous globalization and industrialization have ensured metals are an increasing aspect of daily life. Their usefulness in manufacturing has made them vital to national commerce, security and global economy. However, excess exposure to metals, particularly as a result of environmental contamination or occupational exposures, has been detrimental to overall health. Excess exposure to several metals is considered environmental risk in the aetiology of several neurological and neurodegenerative diseases. Metal-induced neurotoxicity has been a major health concern globally with intensive research to unravel the mechanisms associated with it. Recently, greater focus has been directed at epigenetics to better characterize the underlying mechanisms of metal-induced neurotoxicity. Epigenetic changes are those modifications on the DNA that can turn genes on or off without altering the DNA sequence. This review discusses how epigenetic changes such as DNA methylation, post translational histone modification and noncoding RNA-mediated gene silencing mediate the neurotoxic effects of several metals, focusing on manganese, arsenic, nickel, cadmium, lead, and mercury.
Collapse
Affiliation(s)
- Omamuyovwi M Ijomone
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria.
| | - Olayemi K Ijomone
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria; Department of Anatomy, University of Medical Sciences, Ondo, Nigeria
| | - Joy D Iroegbu
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Chibuzor W Ifenatuoha
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Nzube F Olung
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Michael Aschner
- Departments of Molecular Pharmacology and Neurosciences, Albert Einstein College of Medicine, NY, USA.
| |
Collapse
|
62
|
Silencing of long non-coding RNA LINC01270 inhibits esophageal cancer progression and enhances chemosensitivity to 5-fluorouracil by mediating GSTP1methylation. Cancer Gene Ther 2020; 28:471-485. [PMID: 33199829 DOI: 10.1038/s41417-020-00232-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 08/28/2020] [Accepted: 09/21/2020] [Indexed: 12/17/2022]
Abstract
Esophageal cancer (EC) is a serious digestive malignancy which remains the sixth leading cause of cancer-related deaths worldwide. Emerging evidence suggests the involvement of long non-coding RNAs (lncRNAs) in the tumorigenesis of EC and thus, in this study we explored the potential effects of lncRNA LINC01270 on EC cell proliferation, migration, invasion and, drug resistance via regulation of glutathione S-transferase P1 (GSTP1) methylation. First, we screened out the EC-related differentially expressed lncRNAs, and the expression of our top candidate LINC01270 was quantified in EC tissues and cells. To define the role of LINC01270 in EC progression, we evaluated the proliferation, migration and invasion of EC cells when the LINC01270 was overexpressed or knocked down, in the presence of the GSTP1 methylation inhibitor SGI-1027 and 5-fluorouracil (5-FU). In addition, interaction between LINC01270 and methylation of the GSTP1 promoter was identified. Finally, we assessed transplantable tumor growth in nude mice. LINC01270 was up-regulated and GSTP1 was down-regulated in EC tissues and cells. Silencing of LINC01270 inhibited migration and invasion, and enhanced the sensitivity of 5-FU in EC cells. We found that LINC01270 recruited the DNA methyltransferases DNMT1, DNMT3A and DNMT3B initiating GSTP1 promoter methylation, thereby leading to the proliferation, migration, invasion and drug resistance of EC cells. Moreover, GSTP1 overexpression was observed to reverse the effects of LINC01270 overexpression on EC cells and their response to 5-FU. Taken together, this study shows that inhibition of LINC01270 can lead to suppression of EC progression via demethylation of GSTP1, highlighting this lncRNA as a potential target for EC treatment.
Collapse
|
63
|
Cheng Y, Saville L, Gollen B, Isaac C, Belay A, Mehla J, Patel K, Thakor N, Mohajerani MH, Zovoilis A. Increased processing of SINE B2 ncRNAs unveils a novel type of transcriptome deregulation in amyloid beta neuropathology. eLife 2020; 9:61265. [PMID: 33191914 PMCID: PMC7717908 DOI: 10.7554/elife.61265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/15/2020] [Indexed: 12/18/2022] Open
Abstract
The functional importance of many non-coding RNAs (ncRNAs) generated by repetitive elements and their connection with pathologic processes remains elusive. B2 RNAs, a class of ncRNAs of the B2 family of SINE repeats, mediate through their processing the transcriptional activation of various genes in response to stress. Here, we show that this response is dysfunctional during amyloid beta toxicity and pathology in the mouse hippocampus due to increased levels of B2 RNA processing, leading to constitutively elevated B2 RNA target gene expression and high Trp53 levels. Evidence indicates that Hsf1, a master regulator of stress response, mediates B2 RNA processing in hippocampal cells and is activated during amyloid toxicity, accelerating the processing of SINE RNAs and gene hyper-activation. Our study reveals that in mouse, SINE RNAs constitute a novel pathway deregulated in amyloid beta pathology, with potential implications for similar cases in the human brain, such as Alzheimer’s disease (AD).
Collapse
Affiliation(s)
- Yubo Cheng
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Canada.,Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, Canada.,Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, Canada.,Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, Canada
| | - Luke Saville
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Canada.,Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, Canada.,Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, Canada.,Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, Canada
| | - Babita Gollen
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Canada.,Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, Canada.,Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, Canada.,Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, Canada
| | - Christopher Isaac
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Canada.,Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, Canada.,Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, Canada.,Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, Canada
| | - Abel Belay
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Canada.,Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, Canada.,Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, Canada.,Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, Canada
| | - Jogender Mehla
- Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, Canada
| | - Kush Patel
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Canada.,Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, Canada.,Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, Canada
| | - Nehal Thakor
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Canada.,Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, Canada.,Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, Canada
| | - Majid H Mohajerani
- Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, Canada
| | - Athanasios Zovoilis
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Canada.,Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, Canada.,Canadian Centre for Behavioral Neuroscience, University of Lethbridge, Lethbridge, Canada.,Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, Canada
| |
Collapse
|
64
|
Nuthikattu S, Milenkovic D, Rutledge JC, Villablanca AC. Sex-Dependent Molecular Mechanisms of Lipotoxic Injury in Brain Microvasculature: Implications for Dementia. Int J Mol Sci 2020; 21:E8146. [PMID: 33142695 PMCID: PMC7663125 DOI: 10.3390/ijms21218146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/16/2020] [Accepted: 10/28/2020] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular risk factors and biologic sex play a role in vascular dementia which is characterized by progressive reduction in cognitive function and memory. Yet, we lack understanding about the role sex plays in the molecular mechanisms whereby lipid stress contributes to cognitive decline. Five-week-old low-density lipoprotein deficient (LDL-R -/-) male and female mice and C57BL/6J wild types (WT) were fed a control or Western Diet for 8 weeks. Differential expression of protein coding and non-protein coding genes (DEG) were determined in laser captured hippocampal microvessels using genome-wide microarray, followed by bioinformatic analysis of gene networks, pathways, transcription factors and sex/gender-based analysis (SGBA). Cognitive function was assessed by Y-maze. Bioinformatic analysis revealed more DEGs in females (2412) compared to males (1972). Hierarchical clusters revealed distinctly different sex-specific gene expression profiles irrespective of diet and genotype. There were also fewer and different biologic responses in males compared to females, as well as different cellular pathways and gene networks (favoring greater neuroprotection in females), together with sex-specific transcription factors and non-protein coding RNAs. Hyperlipidemic stress also resulted in less severe cognitive dysfunction in females. This sex-specific pattern of differential hippocampal microvascular RNA expression might provide therapeutic targets for dementia in males and females.
Collapse
Affiliation(s)
- Saivageethi Nuthikattu
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
| | - Dragan Milenkovic
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
- Université Clermont Auvergne, INRA, UNH, CRNH Auvergne, F-63000 Clermont-Ferrand, France
| | - John C. Rutledge
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
| | - Amparo C. Villablanca
- Division of Cardiovascular Medicine, University of California, Davis, CA 95616, USA; (S.N.); (D.M.); (J.C.R.)
| |
Collapse
|
65
|
Paul S, Bravo Vázquez LA, Pérez Uribe S, Roxana Reyes-Pérez P, Sharma A. Current Status of microRNA-Based Therapeutic Approaches in Neurodegenerative Disorders. Cells 2020; 9:cells9071698. [PMID: 32679881 PMCID: PMC7407981 DOI: 10.3390/cells9071698] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/03/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are a key gene regulator and play essential roles in several biological and pathological mechanisms in the human system. In recent years, plenty of miRNAs have been identified to be involved in the development of neurodegenerative disorders (NDDs), thus making them an attractive option for therapeutic approaches. Hence, in this review, we provide an overview of the current research of miRNA-based therapeutics for a selected set of NDDs, either for their high prevalence or lethality, such as Alzheimer's, Parkinson's, Huntington's, Amyotrophic Lateral Sclerosis, Friedreich's Ataxia, Spinal Muscular Atrophy, and Frontotemporal Dementia. We also discuss the relevant delivery techniques, pertinent outcomes, their limitations, and their potential to become a new generation of human therapeutic drugs in the near future.
Collapse
|
66
|
Improta-Caria AC, Nonaka CKV, Cavalcante BRR, De Sousa RAL, Aras Júnior R, Souza BSDF. Modulation of MicroRNAs as a Potential Molecular Mechanism Involved in the Beneficial Actions of Physical Exercise in Alzheimer Disease. Int J Mol Sci 2020; 21:E4977. [PMID: 32674523 PMCID: PMC7403962 DOI: 10.3390/ijms21144977] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/23/2020] [Accepted: 06/23/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer disease (AD) is one of the most common neurodegenerative diseases, affecting middle-aged and elderly individuals worldwide. AD pathophysiology involves the accumulation of beta-amyloid plaques and neurofibrillary tangles in the brain, along with chronic neuroinflammation and neurodegeneration. Physical exercise (PE) is a beneficial non-pharmacological strategy and has been described as an ally to combat cognitive decline in individuals with AD. However, the molecular mechanisms that govern the beneficial adaptations induced by PE in AD are not fully elucidated. MicroRNAs are small non-coding RNAs involved in the post-transcriptional regulation of gene expression, inhibiting or degrading their target mRNAs. MicroRNAs are involved in physiological processes that govern normal brain function and deregulated microRNA profiles are associated with the development and progression of AD. It is also known that PE changes microRNA expression profile in the circulation and in target tissues and organs. Thus, this review aimed to identify the role of deregulated microRNAs in the pathophysiology of AD and explore the possible role of the modulation of microRNAs as a molecular mechanism involved in the beneficial actions of PE in AD.
Collapse
Affiliation(s)
- Alex Cleber Improta-Caria
- Post-Graduate Program in Medicine and Health, Faculty of Medicine, Federal University of Bahia, Bahia 40110-909, Brazil; (A.C.I.-C.); (R.A.J.)
- University Hospital Professor Edgard Santos, Bahia 40110-909, Brazil
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Bahia 40110-909, Brazil; (C.K.V.N.); (B.R.R.C.)
| | - Carolina Kymie Vasques Nonaka
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Bahia 40110-909, Brazil; (C.K.V.N.); (B.R.R.C.)
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro 20000-000, Brazil
| | - Bruno Raphael Ribeiro Cavalcante
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Bahia 40110-909, Brazil; (C.K.V.N.); (B.R.R.C.)
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro 20000-000, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Bahia 40110-909, Brazil
| | - Ricardo Augusto Leoni De Sousa
- Physiological Science Multicentric Program, Federal University of Valleys´ Jequitinhonha and Mucuri, Minas Gerais 30000-000, Brazil;
| | - Roque Aras Júnior
- Post-Graduate Program in Medicine and Health, Faculty of Medicine, Federal University of Bahia, Bahia 40110-909, Brazil; (A.C.I.-C.); (R.A.J.)
- University Hospital Professor Edgard Santos, Bahia 40110-909, Brazil
| | - Bruno Solano de Freitas Souza
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Bahia 40110-909, Brazil; (C.K.V.N.); (B.R.R.C.)
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro 20000-000, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Bahia 40110-909, Brazil
| |
Collapse
|
67
|
Singh T, Yadav S. Role of microRNAs in neurodegeneration induced by environmental neurotoxicants and aging. Ageing Res Rev 2020; 60:101068. [PMID: 32283224 DOI: 10.1016/j.arr.2020.101068] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 03/02/2020] [Accepted: 04/04/2020] [Indexed: 02/06/2023]
Abstract
The progressive loss of neuronal structure and functions resulting in the death of neurons is considered as neurodegeneration. Environmental toxicants induced degeneration of neurons is accelerated with aging. In adult brains, most of the neurons are post-mitotic, and their loss results in the development of diseases like amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), Alzheimer's disease (AD), and Huntington's disease (HD). Neurodegenerative diseases have several similarities at the sub-cellular and molecular levels, such as synaptic degeneration, oxidative stress, inflammation, and cognitive decline, which are also known in brain aging. Identification of these similarities at the molecular level offers hope for the development of new therapeutics to ameliorate all neurodegenerative diseases simultaneously. Aging is known as the most strongly associated additive factor in the pathogenesis of neurodegenerative diseases. Studies carried out so far identified several genes, which are responsible for selective degeneration of neurons in different neurodegenerative diseases. Countless efforts have been made in identifying therapeutics for neurodegenerative diseases; however, the discovery of effective therapy remains elusive. Findings made in the last two decades identified microRNAs (miRNAs) as the most potent post-transcription regulatory RNA molecule, which can condition protein levels in the cell and tissue-specific manner. Identification of miRNAs, which regulate both neurotoxicant and aging-associated degeneration of brain cells, raises the possibility that roads leading to aging and neurotoxicant induced neurodegeneration cross at some point. Identification of miRNAs, which are common to aging and neurotoxicant induced neurodegeneration, will help in understanding the complex mechanism of neurodegenerative disease development. In the future, the use of natural miRNAs in vivo in therapy will be able to tackle several issues of aging and neurodegeneration. In the present review, we have provided a summary of findings made on the role of miRNAs in neurodegeneration and explored the common link made by miRNAs between aging and neurotoxicants induced neurodegeneration.
Collapse
Affiliation(s)
- Tanisha Singh
- Developmental Toxicology Division, Systems Toxicology and Health Risk Assessment Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan,31 Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India; Department of Neurological Surgery, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, Pennsylvania-15213, USA.
| | - Sanjay Yadav
- Developmental Toxicology Division, Systems Toxicology and Health Risk Assessment Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan,31 Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India; Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Raebareli, Munsiganj, Raebareli 229405, UP, India.
| |
Collapse
|
68
|
Brain microRNAs dysregulation: Implication for missplicing and abnormal post-translational modifications of tau protein in Alzheimer’s disease and related tauopathies. Pharmacol Res 2020; 155:104729. [DOI: 10.1016/j.phrs.2020.104729] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 02/01/2020] [Accepted: 02/26/2020] [Indexed: 12/16/2022]
|
69
|
Zimmer AS, Steinberg SM, Smart DD, Gilbert MR, Armstrong TS, Burton E, Houston N, Biassou N, Gril B, Brastianos PK, Carter S, Lyden D, Lipkowitz S, Steeg PS. Temozolomide in secondary prevention of HER2-positive breast cancer brain metastases. Future Oncol 2020; 16:899-909. [PMID: 32270710 PMCID: PMC7270957 DOI: 10.2217/fon-2020-0094] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 03/25/2020] [Indexed: 01/11/2023] Open
Abstract
Brain metastases occur in up to 25-55% of patients with metastatic HER2-positive breast cancer. Standard treatment has high rates of recurrence or progression, limiting survival and quality of life in most patients. Temozolomide (TMZ) is known to penetrate the blood-brain barrier and is US FDA approved for treatment of glioblastoma. Our group has demonstrated that low doses of TMZ administered in a prophylactic, metronomic fashion can significantly prevent development of brain metastases in murine models of breast cancer. Based on these findings, we initiated a secondary-prevention clinical trial with oral TMZ given to HER2-positive breast cancer patients with brain metastases after recent local treatment in combination with T-DM1 for systemic control of disease. Primary end point is freedom from new brain metastases at 1 year. (NCT03190967).
Collapse
Affiliation(s)
- Alexandra S Zimmer
- Women's Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| | - Seth M Steinberg
- Biostatistics & Data Management Section, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| | - Dee Dee Smart
- Radiation Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| | - Terri S Armstrong
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| | - Eric Burton
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| | - Nicole Houston
- Women's Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| | - Nadia Biassou
- Neuro-Radiology, Clinical Center, NIH, Bethesda, MD 20814, USA
| | - Brunilde Gril
- Women's Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| | - Priscilla K Brastianos
- Central Nervous System Metastases Program, Massachusetts General Hospital/Harvard Cancer Center Boston, MA 02114, USA
| | - Scott Carter
- Biostatistics and Computation Biology, Dana-Farber Cancer Institute, Boston, MA 02114, USA
| | - David Lyden
- Pediatric Hematology Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Stanley Lipkowitz
- Women's Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| | - Patricia S Steeg
- Women's Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| |
Collapse
|
70
|
Mo Y, Liu B, Qiu S, Wang X, Zhong L, Han X, Mi F. Down‐regulation of microRNA‐34c‐5p alleviates neuropathic pain
via
the SIRT1/STAT3 signaling pathway in rat models of chronic constriction injury of sciatic nerve. J Neurochem 2020; 154:301-315. [PMID: 32126145 DOI: 10.1111/jnc.14998] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 12/27/2022]
Affiliation(s)
- Yanshuai Mo
- Department of Anesthesiology Linyi People’s Hospital Linyi P.R. China
| | - Benjuan Liu
- Department of Anesthesiology Linyi People’s Hospital Linyi P.R. China
| | - Shuang Qiu
- Department of Anesthesiology Linyi People’s Hospital Linyi P.R. China
| | - Xueqin Wang
- Department of Anesthesiology Linyi People’s Hospital Linyi P.R. China
| | - Lina Zhong
- Department of Anesthesiology Linyi People’s Hospital Linyi P.R. China
| | - Xiao Han
- Department of Anesthesiology Linyi People’s Hospital Linyi P.R. China
| | - Fuli Mi
- Department of Anesthesiology Linyi People’s Hospital Linyi P.R. China
| |
Collapse
|
71
|
Kelly J, Moyeed R, Carroll C, Luo S, Li X. Genetic networks in Parkinson's and Alzheimer's disease. Aging (Albany NY) 2020; 12:5221-5243. [PMID: 32205467 PMCID: PMC7138567 DOI: 10.18632/aging.102943] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/09/2020] [Indexed: 12/14/2022]
Abstract
Parkinson’s disease (PD) and Alzheimer’s disease (AD) are the most common neurodegenerative diseases and there is increasing evidence that they share common physiological and pathological links. Here we have conducted the largest network analysis of PD and AD based on their gene expressions in blood to date. We identified modules that were not preserved between disease and healthy control (HC) networks, and important hub genes and transcription factors (TFs) in these modules. We highlighted that the PD module not preserved in HCs was associated with insulin resistance, and HDAC6 was identified as a hub gene in this module which may have the role of influencing tau phosphorylation and autophagic flux in neurodegenerative disease. The AD module associated with regulation of lipolysis in adipocytes and neuroactive ligand-receptor interaction was not preserved in healthy and mild cognitive impairment networks and the key hubs TRPC5 and BRAP identified as potential targets for therapeutic treatments of AD. Our study demonstrated that PD and AD share common disrupted genetics and identified novel pathways, hub genes and TFs that may be new areas for mechanistic study and important targets in both diseases.
Collapse
Affiliation(s)
- Jack Kelly
- Faculty of Health: Medicine, Dentistry and Human Sciences, Plymouth University, Plymouth PL6 8BU, UK
| | - Rana Moyeed
- Faculty of Science and Engineering, Plymouth University, Plymouth PL6 8BU, UK
| | - Camille Carroll
- Faculty of Health: Medicine, Dentistry and Human Sciences, Plymouth University, Plymouth PL6 8BU, UK
| | - Shouqing Luo
- Faculty of Health: Medicine, Dentistry and Human Sciences, Plymouth University, Plymouth PL6 8BU, UK
| | - Xinzhong Li
- School of Science, Engineering and Design, Teesside University, Middlesbrough TS1 3BX, UK
| |
Collapse
|
72
|
Hu J, Cui F, Zhang X. Morphine modulates hippocampal neurogenesis and contextual memory extinction via miR-34c/Notch1 pathway in male ICR mice. Open Life Sci 2020. [DOI: 10.1515/biol-2020-0006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AbstractBackgroundThe opioid Morphine is known to affect neurogenesis in the hippocampus. Evidence has shown that several microRNAs modulate morphine-induced neurogenesis, and hence morphine-induced contextual memory. This complex network has yet to be elucidated. In this study, we screened for morphine addiction related microRNA and determined its effects on hippocampal neurogenesis and morphine-induced contextual memory using the conditioned place preference (CPP) model.MethodsThe previously established CPP model was utilized in this study. For differential expression of miRNA in the hippocampus, the GeneChip miRNA array was used. Lentivirus technology was used to overexpress or downregulate the miRNA, and changes in expression level was verified with qRT-PCR. Protein expression levels were measured with western blot. Immunofluorescence was used to observe the protein expression during the differentiation of NSCs.ResultsThe results showed that morphine administration upregulated microRNA-34c (miR-34c) and Notch1. Downregulating miR-34c in vivo decreased Notch1 expression and partially rescued the morphine-induced inhibition of the differentiation of neural stem cells (NSCs). This did not affect the morphine-induced proliferation of cells. Furthermore, downregulating miR-34c in vivo prolonged the extinction of morphine-induced contextual memory without affecting acquired CPP response.ConclusionThe miR-34c regulates the hippocampal neurogenesis in addicted mice by up-regulating Notch1 expression, by inhibiting differentiation of neural precursor cells. The miR-34c/Notch1 pathway may be a new potential target for the prevention and treatment of opioid psychotic dependence.
Collapse
Affiliation(s)
- JieWei Hu
- College of Tropical Medicine and Public Health, Second Military Medical University, Shanghai, China
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - FuHua Cui
- College of Tropical Medicine and Public Health, Second Military Medical University, Shanghai, China
- PLA Urumqi General Hospital, Urumqi, Xinjiang, China
| | - XiaoDong Zhang
- College of Tropical Medicine and Public Health, Second Military Medical University, Shanghai, China
| |
Collapse
|
73
|
Bertogliat MJ, Morris-Blanco KC, Vemuganti R. Epigenetic mechanisms of neurodegenerative diseases and acute brain injury. Neurochem Int 2020; 133:104642. [PMID: 31838024 PMCID: PMC8074401 DOI: 10.1016/j.neuint.2019.104642] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/25/2019] [Accepted: 12/09/2019] [Indexed: 12/22/2022]
Abstract
Epigenetic modifications are emerging as major players in the pathogenesis of neurodegenerative disorders and susceptibility to acute brain injury. DNA and histone modifications act together with non-coding RNAs to form a complex gene expression machinery that adapts the brain to environmental stressors and injury response. These modifications influence cell-level operations like neurogenesis and DNA repair to large, intricate processes such as brain patterning, memory formation, motor function and cognition. Thus, epigenetic imbalance has been shown to influence the progression of many neurological disorders independent of aberrations in the genetic code. This review aims to highlight ways in which epigenetics applies to several commonly researched neurodegenerative diseases and forms of acute brain injury as well as shed light on the benefits of epigenetics-based treatments.
Collapse
Affiliation(s)
- Mario J Bertogliat
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Kahlilia C Morris-Blanco
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; William S. Middleton VA Hospital, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; William S. Middleton VA Hospital, Madison, WI, USA.
| |
Collapse
|
74
|
Bas-Orth C, Koch M, Lau D, Buchthal B, Bading H. A microRNA signature of toxic extrasynaptic N-methyl-D-aspartate (NMDA) receptor signaling. Mol Brain 2020; 13:3. [PMID: 31924235 PMCID: PMC6954508 DOI: 10.1186/s13041-020-0546-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/05/2020] [Indexed: 12/27/2022] Open
Abstract
The cellular consequences of N-Methyl-D-Aspartate receptor (NMDAR) stimulation depend on the receptors' subcellular localization. Synaptic NMDARs promote plasticity and survival whereas extrasynaptic NMDARs mediate excitotoxicity and contribute to cell death in neurodegenerative diseases. The mechanisms that couple activation of extrasynaptic NMDARs to cell death remain incompletely understood. We here show that activation of extrasynaptic NMDARs by bath application of NMDA or L-glutamate leads to the upregulation of a group of 19 microRNAs in cultured mouse hippocampal neurons. In contrast, none of these microRNAs is induced upon stimulation of synaptic activity. Increased microRNA expression depends on the pri-miRNA processing enzyme Drosha, but not on de novo gene transcription. These findings suggest that toxic NMDAR signaling involves changes in the expression levels of particular microRNAs.
Collapse
Affiliation(s)
- Carlos Bas-Orth
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany. .,Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120, Heidelberg, Germany.
| | - Mirja Koch
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - David Lau
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Bettina Buchthal
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120, Heidelberg, Germany
| |
Collapse
|
75
|
Sun N, Yang C, He X, Liu Z, Liu S, Li X, Wang Y, Jin R, Zhang K. Impact of Expression and Genetic Variation of microRNA-34b/c on Cognitive Dysfunction in Patients with Major Depressive Disorder. Neuropsychiatr Dis Treat 2020; 16:1543-1554. [PMID: 32606706 PMCID: PMC7311205 DOI: 10.2147/ndt.s247787] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/17/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Patients suffering from major depressive disorder (MDD) commonly demonstrate lower performance across multiple cognitive domains. Cognitive impairment is an intrinsic characteristic of MDD status and is often influenced by genetic factors. microRNAs (miRNAs or miRs) have been shown to have important implications in the etiology of MDD. Therefore, we aimed to identify and analyze the impact of expression and genetic variation of miR-34b/c on cognitive dysfunction in MDD. METHODS First, we analyzed miR-34c-5p expression in 48 cases of MDD and 54 healthy controls in a Chinese population using qRT-PCR. We assessed the relationship between the level of miR-34c-5p expression and cognitive performance by Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) and the Trail Making Test (TMT). Second, in order to characterize allelic effects of miR-34b/c on cognitive performance in MDD patients, we performed genetic association analysis of single-nucleotide polymorphism (SNP) loci of the MIR34B/C genes with cognitive function in a second group consisting of 531 MDD patients and 267 healthy controls. RESULTS We found a significant negative correlation between the level of miR-34c-5p expression and both the language and delayed memory index scores in patients with MDD. We also found a significant positive correlation between the level of miR-34c-5p expression and the time required to complete tests A and B of the TMT. The interaction between the rs2187473 genotype and the disease was significant for both immediate memory and delayed memory. In the patient group, the rs2187473 CC genotype was significantly associated with higher performance on immediate memory (F = 6.683, p < 0.05) and delayed memory tasks (F = 4.221, p < 0.05). CONCLUSION Our findings suggest that changes in miR-34c expression level have important impacts on cognitive function in patients with MDD. In particular, the polymorphism rs2187473 is a potential genetic risk factor for cognitive function in MDD, which may be of clinical use.
Collapse
Affiliation(s)
- Ning Sun
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.,Nursing College of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Chunxia Yang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xiaoting He
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.,University-Town Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhifen Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Sha Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xinrong Li
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Yanfang Wang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Ruihua Jin
- Nursing College of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Kerang Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| |
Collapse
|
76
|
MicroRNA-34a Acutely Regulates Synaptic Efficacy in the Adult Dentate Gyrus In Vivo. Mol Neurobiol 2019; 57:1432-1445. [PMID: 31754996 DOI: 10.1007/s12035-019-01816-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/11/2019] [Indexed: 12/30/2022]
Abstract
Activity-dependent synaptic plasticity involves rapid regulation of neuronal protein synthesis on a time-scale of minutes. miRNA function in synaptic plasticity and memory formation has been elucidated by stable experimental manipulation of miRNA expression and activity using transgenic approaches and viral vectors. However, the impact of rapid miRNA modulation on synaptic efficacy is unknown. Here, we examined the effect of acute (12 min), intrahippocampal infusion of a miR-34a antagonist (antimiR) on medial perforant path-evoked synaptic transmission in the dentate gyrus of adult anesthetised rats. AntimiR-34a infusion acutely depressed medial perforant path-evoked field excitatory post-synaptic potentials (fEPSPs). The fEPSP decrease was detected within 9 min of infusion, lasted for hours, and was associated with knockdown of antimiR-34a levels. AntimiR-34a-induced synaptic depression was sequence-specific; no changes were elicited by infusion of scrambled or mismatch control. The rapid modulation suggests that a target, or set of targets, is regulated by miR-34a. Western blot analysis of dentate gyrus lysates revealed enhanced expression of Arc, a known miR-34a target, and four novel predicted targets (Ctip2, PKI-1α, TCF4 and Ube2g1). Remarkably, antimiR-34a had no effect when infused during the maintenance phase of long-term potentiation. We conclude that miR-34a regulates basal synaptic efficacy in the adult dentate gyrus in vivo. To our knowledge, these in vivo findings are the first to demonstrate acute (< 9 min) regulation of synaptic efficacy in the adult brain by a miRNA.
Collapse
|
77
|
Jain G, Stuendl A, Rao P, Berulava T, Pena Centeno T, Kaurani L, Burkhardt S, Delalle I, Kornhuber J, Hüll M, Maier W, Peters O, Esselmann H, Schulte C, Deuschle C, Synofzik M, Wiltfang J, Mollenhauer B, Maetzler W, Schneider A, Fischer A. A combined miRNA-piRNA signature to detect Alzheimer's disease. Transl Psychiatry 2019; 9:250. [PMID: 31591382 PMCID: PMC6779890 DOI: 10.1038/s41398-019-0579-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/05/2019] [Accepted: 08/18/2019] [Indexed: 01/03/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder causing huge emotional and economic burden to our societies. An effective therapy has not been implicated yet, which is in part also due to the fact that pathological changes occur years before clinical symptoms manifest. Thus, there is a great need for the development of a translatable biomarker. Recent evidence highlights microRNAs as candidate biomarkers. In this study, we use next-generation sequencing to study the small noncoding RNAome (sncRNAome) in exosomes derived from human cerebrospinal fluid (CSF). We show that the sncRNAome from CSF-derived exosomes is dominated not only by microRNAs (miRNAs) but also by PIWI-interacting RNAs (piRNAs). We define a combined signature consisting of three miRNAs and three piRNAs that are suitable to detect AD with an AUC of 0.83 in a replication cohort and furthermore predict the conversion of mild-cognitive impaired (MCI) patients to AD dementia with an AUC of 0.86 for the piRNA signature. When combining the smallRNA signature with pTau and Aβ 42/40 ratio the AUC reaches 0.98. Our study reports a novel exosomal small noncoding RNA signature to detect AD pathology and provides the first evidence that in addition to miRNAs, piRNAs should also be considered as a candidate biomarker for AD.
Collapse
Affiliation(s)
- Gaurav Jain
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany
| | - Anne Stuendl
- 0000 0004 0438 0426grid.424247.3Translational Dementia Research, German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Pooja Rao
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany
| | - Tea Berulava
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany
| | - Tonatiuh Pena Centeno
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany ,Bioinformatics Unit, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Goettingen, Germany
| | - Lalit Kaurani
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany
| | - Susanne Burkhardt
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany
| | - Ivana Delalle
- 0000 0004 0367 5222grid.475010.7Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Johannes Kornhuber
- 0000 0001 2107 3311grid.5330.5Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Michael Hüll
- 0000 0000 9428 7911grid.7708.8Center for Geriatric Medicine and Gerontology, University Medical Center Freiburg, 79106 Freiburg, Germany ,0000 0000 9428 7911grid.7708.8Department of Psychiatry and Psychotherapy, University Medical Centre Freiburg, 79106 Freiburg, Germany
| | - Wolfgang Maier
- 0000 0001 2240 3300grid.10388.32Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, 53127 Bonn, Germany
| | - Oliver Peters
- 0000 0001 2248 7639grid.7468.dDepartment of Psychiatry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 12200 Berlin, Germany ,0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), 12203 Berlin, Germany ,0000 0001 1014 0849grid.419491.0Memory Clinic and Dementia Prevention Center, Experimental and Clinical Research Center (ECRC), 13125 Berlin, Germany
| | - Hermann Esselmann
- 0000 0001 0482 5331grid.411984.1Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), 37075 Göttingen, Germany
| | - Claudia Schulte
- 0000 0001 2190 1447grid.10392.39Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany ,0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), 72076 Tuebingen, Germany
| | - Christian Deuschle
- 0000 0001 2190 1447grid.10392.39Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany ,0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), 72076 Tuebingen, Germany
| | - Mathis Synofzik
- 0000 0001 2190 1447grid.10392.39Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany ,0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), 72076 Tuebingen, Germany
| | - Jens Wiltfang
- 0000 0001 0482 5331grid.411984.1Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), 37075 Göttingen, Germany ,0000000123236065grid.7311.4iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal ,German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany
| | - Brit Mollenhauer
- 0000 0001 0482 5331grid.411984.1Department of Neurology, University Medical Center Göttingen (UMG), 37075 Göttingen, Germany ,grid.440220.0Paracelsus-Elena-Klinik, 34128 Kassel, Germany
| | - Walter Maetzler
- 0000 0001 2190 1447grid.10392.39Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany ,0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), 72076 Tuebingen, Germany ,0000 0004 0646 2097grid.412468.dDepartment of Neurology, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Anja Schneider
- Translational Dementia Research, German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany. .,Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, 53127, Bonn, Germany.
| | - Andre Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075, Göttingen, Germany. .,Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), 37075, Göttingen, Germany.
| |
Collapse
|
78
|
MiR-124 suppression in the prefrontal cortex reduces depression-like behavior in mice. Biosci Rep 2019; 39:BSR20190186. [PMID: 31431514 PMCID: PMC6744582 DOI: 10.1042/bsr20190186] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/30/2019] [Accepted: 08/19/2019] [Indexed: 02/04/2023] Open
Abstract
Depression is a potentially life-threatening mental disorder with unknown etiology. Several microRNAs (miRNAs) have been shown to play critical roles in the etiology of depression. Here, we aim to elucidate the anti-depressive behavior of miR-124 suppression in prefrontal cortex (PFC). Quantitative real-time PCR (RT-PCR) was used to evaluate the expression of miR-124 and SIRT1 in the PFC of a chronic unpredictable mild stress (CUMS) model. The PFC of C57BL/6J mice was bilaterally injected with lentiviral vectors (LV) for ectopic expression of SIRT1, miR-124, or miR-124 inhibitor (si-miR-124). The anti-depressive behavior was observed after injection of LV-SIRT1 or LV-si-miR-124 into the PFC, using behavior tests including latency to feed, food and water intake, sucrose preference test, and forced swimming test. MiR-124 overexpression and inhibition resulted in upregulation and down-regulation of SIRT1 and cyclic AMP responsive element binding protein 1 (CREB1), respectively. MiR-124 overexpression exacerbated depression-like behaviors and decreased SIRT1. Further, dual-luciferase assay confirmed that SIRT1 was a target of miR-124. Taken together, a potential molecular regulation of miR-124 on SIRT1 is revealed by our study and miR-124 suppression in PFC is a potential strategy to reduce depression-like behavior.
Collapse
|
79
|
Lo Iacono L, Ielpo D, Accoto A, Di Segni M, Babicola L, D’Addario SL, Ferlazzo F, Pascucci T, Ventura R, Andolina D. MicroRNA-34a Regulates the Depression-like Behavior in Mice by Modulating the Expression of Target Genes in the Dorsal Raphè. Mol Neurobiol 2019; 57:823-836. [DOI: 10.1007/s12035-019-01750-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 08/26/2019] [Indexed: 01/06/2023]
|
80
|
Kiss T, Giles CB, Tarantini S, Yabluchanskiy A, Balasubramanian P, Gautam T, Csipo T, Nyúl-Tóth Á, Lipecz A, Szabo C, Farkas E, Wren JD, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) supplementation promotes anti-aging miRNA expression profile in the aorta of aged mice, predicting epigenetic rejuvenation and anti-atherogenic effects. GeroScience 2019; 41:419-439. [PMID: 31463647 PMCID: PMC6815288 DOI: 10.1007/s11357-019-00095-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 08/21/2019] [Indexed: 12/11/2022] Open
Abstract
Understanding molecular mechanisms involved in vascular aging is essential to develop novel interventional strategies for treatment and prevention of age-related vascular pathologies. Recent studies provide critical evidence that vascular aging is characterized by NAD+ depletion. Importantly, in aged mice, restoration of cellular NAD+ levels by treatment with the NAD+ booster nicotinamide mononucleotide (NMN) exerts significant vasoprotective effects, improving endothelium-dependent vasodilation, attenuating oxidative stress, and rescuing age-related changes in gene expression. Strong experimental evidence shows that dysregulation of microRNAs (miRNAs) has a role in vascular aging. The present study was designed to test the hypothesis that age-related NAD+ depletion is causally linked to dysregulation of vascular miRNA expression. A corollary hypothesis is that functional vascular rejuvenation in NMN-treated aged mice is also associated with restoration of a youthful vascular miRNA expression profile. To test these hypotheses, aged (24-month-old) mice were treated with NMN for 2 weeks and miRNA signatures in the aortas were compared to those in aortas obtained from untreated young and aged control mice. We found that protective effects of NMN treatment on vascular function are associated with anti-aging changes in the miRNA expression profile in the aged mouse aorta. The predicted regulatory effects of NMN-induced differentially expressed miRNAs in aged vessels include anti-atherogenic effects and epigenetic rejuvenation. Future studies will uncover the mechanistic role of miRNA gene expression regulatory networks in the anti-aging effects of NAD+ booster treatments and determine the links between miRNAs regulated by NMN and sirtuin activators and miRNAs known to act in the conserved pathways of aging and major aging-related vascular diseases.
Collapse
Affiliation(s)
- Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics / Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Cory B Giles
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Oklahoma Medical Research Foundation, Genes & Human Disease Research Program, Oklahoma City, OK and Department of Biochemistry and Molecular Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
| | - Tripti Gautam
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
| | - Tamas Csipo
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics / Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
- Department of Public Health / Doctoral School of Basic and Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Institute of Biophysics, Biological Research Centre / Theoretical Medicine Doctoral School, Hungarian Academy of Sciences, Szeged, Hungary
| | - Agnes Lipecz
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics / Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
- Department of Public Health / Doctoral School of Basic and Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Csaba Szabo
- Chair of Pharmacology, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Eszter Farkas
- Department of Medical Physics and Informatics / Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Jonathan D Wren
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Oklahoma Medical Research Foundation, Genes & Human Disease Research Program, Oklahoma City, OK and Department of Biochemistry and Molecular Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics / Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Institute of Human Physiology and Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA.
- Department of Medical Physics and Informatics / Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary.
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- Department of Public Health / Doctoral School of Basic and Translational Medicine, Semmelweis University, Budapest, Hungary.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
81
|
Kinser HE, Pincus Z. MicroRNAs as modulators of longevity and the aging process. Hum Genet 2019; 139:291-308. [PMID: 31297598 DOI: 10.1007/s00439-019-02046-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/05/2019] [Indexed: 12/22/2022]
Abstract
MicroRNAs (miRNAs) are short, non-coding RNAs that post-transcriptionally repress translation or induce mRNA degradation of target transcripts through sequence-specific binding. miRNAs target hundreds of transcripts to regulate diverse biological pathways and processes, including aging. Many microRNAs are differentially expressed during aging, generating interest in their use as aging biomarkers and roles as regulators of the aging process. In the invertebrates Caenorhabditis elegans and Drosophila, a number of miRNAs have been found to both positive and negatively modulate longevity through canonical aging pathways. Recent studies have also shown that miRNAs regulate age-associated processes and pathologies in a diverse array of mammalian tissues, including brain, heart, bone, and muscle. The review will present an overview of these studies, highlighting the role of individual miRNAs as biomarkers of aging and regulators of longevity and tissue-specific aging processes.
Collapse
Affiliation(s)
- Holly E Kinser
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, USA
| | - Zachary Pincus
- Department of Developmental Biology and Department of Genetics, Washington University in St. Louis, St. Louis, USA.
| |
Collapse
|
82
|
Over-expression of miR-34a induces rapid cognitive impairment and Alzheimer's disease-like pathology. Brain Res 2019; 1721:146327. [PMID: 31295467 DOI: 10.1016/j.brainres.2019.146327] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/28/2019] [Accepted: 07/07/2019] [Indexed: 01/24/2023]
Abstract
Autosomal dominant Alzheimer disease (AD) is caused by rare mutations in one of three specific genes. This is in contrast to idiopathic, late-onset AD (LOAD), which has a more polygenetic risk profile and represents more than 95% of cases. Previously, we have demonstrated that increased expression of microRNA (miRNA)-34a (miR-34a) in AD brain targets genes linked to synaptic plasticity, energy metabolism, and resting state network activity. Here we report the generation of a heterozygous, conditional miR-34a overexpression mouse (miR-34a+/-(TetR-TetO-miR-34a) Transgenic Mice). Doxycycline-treated mice of either sex exhibited profound behavioral impairment compared to untreated groups with only 1-2 months of over-expression of miR-34a. Cognitive impairment of individual mice in T- and Y-maze tasks correlated with elevated miR-34a expression in many parts of the brain including the hippocampus and prefrontal cortex, regions which are known to be involved in this task and implicated in LOAD dysfunction. Immunocytochemistry of brain sections from mice show high amyloid β and phosphorylated tau-specific staining in the hippocampus and cortex. Analysis of protein samples from these mice revealed that miR-34a targets specific genes involved in memory formation, amyloid precursor protein (APP) metabolism and phosphorylation-dephosphorylation of tau. Thus, our results suggest that the polygenetic dysfunction caused by miR-34a may occur in LOAD and disclose miR-34a as a potential therapeutic target. SIGNIFICANCE STATEMENT: Late-onset Alzheimer disease (LOAD) is associated with multiple gene alleles, a polygenetic profile of risk factors that is difficult to model in animals. Our approach to modeling LOAD was to produce a conditional over-expressing, miR-34a mouse using doxycycline-induction to activate expression. We observed that miR-34a over-expression results in a rapid cognitive impairment, associated with accumulation of intracellular Aβ and tau hyperphosphorylation in multiple brain regions. Targets for miR-34a, including ADAM10, NMDAR 2B, and SIRT1 RNAs, were profoundly reduced by miR-34a over-expression. Collectively, these results indicate that a rapid, profound cognitive decline and Alzheimer's disease neuropathology can be induced with miR-34a over-expression, suggesting that this animal model may represent a polygenetic risk factor model for LOAD.
Collapse
|
83
|
Su L, Chen S, Zheng C, Wei H, Song X. Meta-Analysis of Gene Expression and Identification of Biological Regulatory Mechanisms in Alzheimer's Disease. Front Neurosci 2019; 13:633. [PMID: 31333395 PMCID: PMC6616202 DOI: 10.3389/fnins.2019.00633] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/31/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD), also known as senile dementia, is a progressive neurodegenerative disease. The etiology and pathogenesis of AD have not yet been elucidated. We examined common differentially expressed genes (DEGs) from different AD tissue microarray datasets by meta-analysis and screened the AD-associated genes from the common DEGs using GCBI. Then we studied the gene expression network using the STRING database and identified the hub genes using Cytoscape. Furthermore, we analyzed the microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and single nucleotide polymorphisms (SNPs) associated with the AD-associated genes, and then identified feed-forward loops. Finally, we performed SNP analysis of the AD-associated genes. Our results identified 207 common DEGs, of which 57 have previously been reported to be associated with AD. The common DEG expression network identified eight hub genes, all of which were previously known to be associated with AD. Further study of the regulatory miRNAs associated with the AD-associated genes and other genes specific to neurodegenerative diseases revealed 65 AD-associated miRNAs. Analysis of the miRNA associated transcription factor-miRNA-gene-gene associated TF (mTF-miRNA-gene-gTF) network around the AD-associated genes revealed 131 feed-forward loops (FFLs). Among them, one important FFL was found between the gene SERPINA3, hsa-miR-27a, and the transcription factor MYC. Furthermore, SNP analysis of the AD-associated genes identified 173 SNPs, and also found a role in AD for miRNAs specific to other neurodegenerative diseases, including hsa-miR-34c, hsa-miR-212, hsa-miR-34a, and hsa-miR-7. The regulatory network constructed in this study describes the mechanism of cell regulation in AD, in which miRNAs and lncRNAs can be considered AD regulatory factors.
Collapse
Affiliation(s)
- Lining Su
- Department of Basic Medicine, Hebei North University, Zhangjiakou, China
| | - Sufen Chen
- Institute of Educational Science, Zhangjiakou, China
| | | | - Huiping Wei
- Department of Basic Medicine, Hebei North University, Zhangjiakou, China
| | - Xiaoqing Song
- Department of Basic Medicine, Hebei North University, Zhangjiakou, China
| |
Collapse
|
84
|
Marangon D, Raffaele S, Fumagalli M, Lecca D. MicroRNAs change the games in central nervous system pharmacology. Biochem Pharmacol 2019; 168:162-172. [PMID: 31251938 DOI: 10.1016/j.bcp.2019.06.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/20/2019] [Indexed: 01/01/2023]
Abstract
MicroRNAs (miRNAs) represent a class of important post-transcriptional regulators of gene expression, enabling cells to follow their intrinsic developmental program. By directly binding to their targets, miRNAs can both promote transcriptional patterns in crucial steps of cell growth, and act as powerful buffering system that titrate protein content in case of aberrant gene expression. The literature of the last decade showed that the presence of tissue-enriched miRNAs in body fluids could be reminiscent of disease state. This is particularly relevant in neurodegenerative disorders, in which peripheral biomarkers could be helpful means to detect disease onset. However, dysregulation of miRNAs is not merely a consequence of disease, but directly contributes to pathological outcomes. On this basis, increasing interest is growing in the development of pharmacological agents targeting specific miRNAs. Actually, this apparently futuristic approach is already part of the current therapies. In fact, several drugs approved for CNS disorders, such as L-Dopa or valproic acid, were also demonstrated to restore some miRNAs. Moreover, ongoing clinical trials demonstrated that miRNA-based drugs are effective against tumors, suggesting that miRNAs also represent a promising class of therapeutic molecules. However, several issues still need to be addressed, particularly in case of CNS diseases, in which stability and delivery are crucial aspects of the therapy. In this commentary, we highlighted potential advantages and limitations of miRNAs as next generation targets in CNS pharmacology, focusing on multiple sclerosis, a chronic demyelinating disease lacking specific therapeutic targets and bona-fide biomarkers.
Collapse
Affiliation(s)
- Davide Marangon
- Laboratorio di Farmacologia Molecolare e Cellulare della Trasmissione Purinergica, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milano, Italy
| | - Stefano Raffaele
- Laboratorio di Farmacologia Molecolare e Cellulare della Trasmissione Purinergica, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milano, Italy
| | - Marta Fumagalli
- Laboratorio di Farmacologia Molecolare e Cellulare della Trasmissione Purinergica, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milano, Italy
| | - Davide Lecca
- Laboratorio di Farmacologia Molecolare e Cellulare della Trasmissione Purinergica, Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milano, Italy.
| |
Collapse
|
85
|
Herrera-Espejo S, Santos-Zorrozua B, Álvarez-González P, Lopez-Lopez E, Garcia-Orad Á. A Systematic Review of MicroRNA Expression as Biomarker of Late-Onset Alzheimer's Disease. Mol Neurobiol 2019; 56:8376-8391. [PMID: 31240600 DOI: 10.1007/s12035-019-01676-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/09/2019] [Indexed: 12/11/2022]
Abstract
Late-onset Alzheimer's disease (LOAD) is a high-occurrence neurological disorder but the difficulty in identifying precise and early biomarkers has complicated the understanding of the disease and the development of new treatments. In this sense, important knowledge is emerging regarding novel molecular and biological candidates with diagnostic potential, including microRNAs (miRNAs), which have a key role in gene repression. The aim of this systematic review was to define the role of miRNAs' expression as biomarkers for LOAD both in brain tissues, which could help understand the biology of the disease, and circulating tissues, which could serve as non-invasive markers of the pathology. A systematic search was performed in Web of Science and PubMed using the keywords ((Alzheimer or Alzheimer's) and (microRNA or microRNAs or miRNA or miRNAs or miR)) until August 2018 to retrieve all articles that presented independent original data evaluating the impact of miRNA expression on the development of LOAD in human population. A total of 90 studies investigating the role of miRNAs' expression in the development of LOAD were identified. While other widely studied miRNAs such as hsa-miR-146a presented contradictory results among studies, deregulation in brain tissue of seven miRNAs, hsa-miR-16-5p, hsa-miR-34a-5p, hsa-miR-107, hsa-miR-125-5p, hsa-miR-132-3p, hsa-miR-181-3p, and hsa-miR-212-3p, was consistently identified in LOAD patients. Their role in the disease could be mediated through the regulation of key pathways, such as axon guidance, longevity, insulin, and MAPK signaling pathway. However, regarding their role as non-invasive biomarkers of LOAD in fluids, although the limited results available are promising, further studies are required.
Collapse
Affiliation(s)
- Soraya Herrera-Espejo
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Medicine and Nursing, University of The Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940, Leioa, Spain
| | - Borja Santos-Zorrozua
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Medicine and Nursing, University of The Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940, Leioa, Spain
| | - Paula Álvarez-González
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Medicine and Nursing, University of The Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940, Leioa, Spain
| | - Elixabet Lopez-Lopez
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Medicine and Nursing, University of The Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940, Leioa, Spain.
- BioCruces Bizkaia Health Research Institute, Barakaldo, Spain.
| | - África Garcia-Orad
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Medicine and Nursing, University of The Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940, Leioa, Spain
- BioCruces Bizkaia Health Research Institute, Barakaldo, Spain
| |
Collapse
|
86
|
Wang T, Li B, Wang Z, Yuan X, Chen C, Zhang Y, Xia Z, Wang X, Yu M, Tao W, Zhang L, Wang X, Zhang Z, Guo X, Ning G, Feng S, Chen X. miR-155-5p Promotes Dorsal Root Ganglion Neuron Axonal Growth in an Inhibitory Microenvironment via the cAMP/PKA Pathway. Int J Biol Sci 2019; 15:1557-1570. [PMID: 31337984 PMCID: PMC6643145 DOI: 10.7150/ijbs.31904] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/29/2019] [Indexed: 12/13/2022] Open
Abstract
Sensory dysfunction post spinal cord injury causes patients great distress. Sciatic nerve conditioning injury (SNCI) has been shown to restore sensory function after spinal cord dorsal column injury (SDCL); however, the underlying mechanism of this recovery remains unclear. We performed a microarray assay to determine the associated miRNAs that might regulate the process of SNCI promoting SDCL repair. In total, 13 miRNAs were identified according to our inclusion criteria, and RT-qPCR was used to verify the microarray results. Among the 13 miRNAs, the miR-155-5p levels were decreased at 9 h, 3 d, 7 d, 14 d, 28 d, 2 m and 3 m timepoints in the SDCL group, while the SNCI group had a smaller decrease. Thus, miR-155-5p was chosen for further study after a literature review and an analysis with the TargetScan online tool. Specifically, miR-155-5p targets PKI-α, and the expression pattern of PKI-α was opposite that of miR-155-5p in both the SDCL and SNCI groups. Interestingly, miR-155-5p could promote dorsal root ganglion (DRG) neuron axon growth via the cAMP/PKA pathway and in a TNF-α, IL-1β or MAG inhibitory microenvironment in vitro. Furthermore, miR-155-5p could regulate the cAMP/PKA pathway and promote sensory conduction function recovery post dorsal column injury as detected by NF-200 immunohistochemistry, somatosensory-evoked potentials, BBB scale and tape removal test. Collectively, our results demonstrated that miR-155-5p participates in the molecular mechanism by which SNCI promotes the repair of SDCL and that upregulated miR-155-5p can repair SDCL by enhancing DRG neuron axon growth via the cAMP/PKA pathway. These findings suggest a novel treatment target for spinal cord injury.
Collapse
Affiliation(s)
- Tianyi Wang
- Department of Orthopedics, The 981st Hospital of the Chinese People's Liberation Army, Chengde 067000, Hebei Province, P.R. China
| | - Bo Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Zhijie Wang
- Department of Pediatric Internal Medicine, Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei Province, P.R. China
| | - Xin Yuan
- Department of Spine Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing 100000, P.R. China
| | - Chuanjie Chen
- Department of Orthopedics, Chengde Central Hospital, Chengde 067000, Hebei Province, P.R. China
| | - Yanjun Zhang
- Department of Spine Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing 100000, P.R. China
| | - Ziwei Xia
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Xin Wang
- Chengde Medical University, Chengde 067000, Hebei Province, P.R. China
| | - Mei Yu
- Leukemia Center, Chinese Academy of Medical Sciences & Peking Union of Medical College, Institute of Hematology & Hospital of Blood Diseases, Tianjin 30020, P.R. China
| | - Wen Tao
- Chengde Medical University, Chengde 067000, Hebei Province, P.R. China
| | - Liang Zhang
- Department of Orthopedics, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Xu Wang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Zheng Zhang
- Department of Orthopedics, The 981st Hospital of the Chinese People's Liberation Army, Chengde 067000, Hebei Province, P.R. China
| | - Xiaoling Guo
- Department of Neurology, The 981st Hospital of the Chinese People's Liberation Army, Chengde 067000, Hebei Province, P.R. China
| | - Guangzhi Ning
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Xueming Chen
- Department of Spine Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing 100000, P.R. China
| |
Collapse
|
87
|
Ge QD, Xie C, Zhang H, Tan Y, Wan CW, Wang WJ, Jin TX. Differential Expression of miRNAs in the Hippocampi of Offspring Rats Exposed to Fluorine Combined with Aluminum during the Embryonic Stage and into Adulthood. Biol Trace Elem Res 2019; 189:463-477. [PMID: 30033483 DOI: 10.1007/s12011-018-1445-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/12/2018] [Indexed: 11/25/2022]
Abstract
A previous study from our team found that continuous exposure to fluorine combined with aluminum (FA) impaired the neurobehavioral reflexes, spatial learning, and memory of offspring rats. To date, the specific mechanisms for these changes are unclear. Here, high-throughput sequencing was utilized to analyze the microRNA (miRNA) profile of the hippocampi in the offspring of rats exposed to FA during the embryonic stage and into adulthood through tap water supplemented with NaF and AlCl3 at concentrations of (0, 0); (60, 600); (120, 600); and (240, 600) mg/L, respectively. qRT-PCR was performed to validate the reliability of the sequence data. Twenty differentially expressed miRNAs were selected for further analysis using bioinformatics tools. Several genes related to neuromodulation were found to be regulated by miR-10a-5p, miR-34b-5p, and miR-182, which might be harmful to normal nerve function. The protein levels of brain-derived neurotrophic factor (BDNF) and tyrosine receptor kinase B (TrkB) in hippocampus were markedly downregulated. These data suggest that miR-10a-5p, miR-34b-5p, and miR-182 and BDNF-TrkB signaling pathway are involved in mechanisms of hippocampal damage in the offspring of rats exposed to FA. HIGHLIGHTS: • Multiple miRNAs were significantly differentially expressed in offspring rat hippocampus after fluorine combined with aluminum (FA) exposure. • Twenty differentially expressed miRNAs might mediate FA-induced developmental neurotoxicity. • MiR-10a-5p, miR-34b-5p, and miR-182 were closely related to neurotoxic signaling of FA. • The BDNF-TrkB learning and memory-associated pathway was downregulated in the hippocampus after FA exposure.
Collapse
Affiliation(s)
- Qi-Di Ge
- Department of Occupational Health and Environmental Hygiene, School of Public Health, Guizhou Medical University, University Town, Guian new district, Guiyang, 550025, Guizhou, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Chun Xie
- Department of Occupational Health and Environmental Hygiene, School of Public Health, Guizhou Medical University, University Town, Guian new district, Guiyang, 550025, Guizhou, China.
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, Guizhou, China.
| | - Hua Zhang
- Department of Occupational Health and Environmental Hygiene, School of Public Health, Guizhou Medical University, University Town, Guian new district, Guiyang, 550025, Guizhou, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Ying Tan
- Department of Occupational Health and Environmental Hygiene, School of Public Health, Guizhou Medical University, University Town, Guian new district, Guiyang, 550025, Guizhou, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Chang-Wu Wan
- Department of Forensic Pathology, School of Forensic Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Wen-Juan Wang
- Department of Occupational Health and Environmental Hygiene, School of Public Health, Guizhou Medical University, University Town, Guian new district, Guiyang, 550025, Guizhou, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Ting-Xu Jin
- Department of Occupational Health and Environmental Hygiene, School of Public Health, Guizhou Medical University, University Town, Guian new district, Guiyang, 550025, Guizhou, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| |
Collapse
|
88
|
Orang AV, Petersen J, McKinnon RA, Michael MZ. Micromanaging aerobic respiration and glycolysis in cancer cells. Mol Metab 2019; 23:98-126. [PMID: 30837197 PMCID: PMC6479761 DOI: 10.1016/j.molmet.2019.01.014] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/22/2019] [Accepted: 01/30/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Cancer cells possess a common metabolic phenotype, rewiring their metabolic pathways from mitochondrial oxidative phosphorylation to aerobic glycolysis and anabolic circuits, to support the energetic and biosynthetic requirements of continuous proliferation and migration. While, over the past decade, molecular and cellular studies have clearly highlighted the association of oncogenes and tumor suppressors with cancer-associated glycolysis, more recent attention has focused on the role of microRNAs (miRNAs) in mediating this metabolic shift. Accumulating studies have connected aberrant expression of miRNAs with direct and indirect regulation of aerobic glycolysis and associated pathways. SCOPE OF REVIEW This review discusses the underlying mechanisms of metabolic reprogramming in cancer cells and provides arguments that the earlier paradigm of cancer glycolysis needs to be updated to a broader concept, which involves interconnecting biological pathways that include miRNA-mediated regulation of metabolism. For these reasons and in light of recent knowledge, we illustrate the relationships between metabolic pathways in cancer cells. We further summarize our current understanding of the interplay between miRNAs and these metabolic pathways. This review aims to highlight important metabolism-associated molecular components in the hunt for selective preventive and therapeutic treatments. MAJOR CONCLUSIONS Metabolism in cancer cells is influenced by driver mutations but is also regulated by posttranscriptional gene silencing. Understanding the nuanced regulation of gene expression in these cells and distinguishing rapid cellular responses from chronic adaptive mechanisms provides a basis for rational drug design and novel therapeutic strategies.
Collapse
Affiliation(s)
- Ayla V Orang
- Flinders Centre for Innovation in Cancer, Flinders University, Flinders Medical Centre, Adelaide, South Australia 5042, Australia.
| | - Janni Petersen
- Flinders Centre for Innovation in Cancer, Flinders University, Flinders Medical Centre, Adelaide, South Australia 5042, Australia.
| | - Ross A McKinnon
- Flinders Centre for Innovation in Cancer, Flinders University, Flinders Medical Centre, Adelaide, South Australia 5042, Australia.
| | - Michael Z Michael
- Flinders Centre for Innovation in Cancer, Flinders University, Flinders Medical Centre, Adelaide, South Australia 5042, Australia.
| |
Collapse
|
89
|
Human Brain Injury and miRNAs: An Experimental Study. Int J Mol Sci 2019; 20:ijms20071546. [PMID: 30934805 PMCID: PMC6479766 DOI: 10.3390/ijms20071546] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/12/2022] Open
Abstract
Brain damage is a complex dysfunction that involves a variety of conditions whose pathogenesis involves a number of mediators that lead to clinical sequelae. For this reason, the identification of specific circulating and/or tissue biomarkers which could indicate brain injury is challenging. This experimental study focused on microRNAs (miRNAs), a well-known diagnostic tool both in the clinical setting and in medico-legal investigation. Previous studies demonstrated that specific miRNAs (miR-21, miR-34, miR-124, miR-132, and miR-200b) control important target genes involved in neuronal apoptosis and neuronal stress-induced adaptation. Thus, in this experimental setting, their expression was evaluated in three selected groups of cadavers: drug abusers (cocaine), ischemic-stroke-related deaths, and aging damage in elder people who died from other neurological causes. The results demonstrated that the drug abuser group showed a higher expression of miR-132 and miR-34, suggesting a specific pathway in consumption-induced neurodegeneration. Instead, miR-200b and miR-21 dysregulation was linked to age-related cognitive impairment, and finally, stroke events and consequences were associated with an alteration in miR-200b, miR-21, and miR-124; significantly higher levels of this last expression are strongly sensitive for ischemic damage. Moreover, these results suggest that these expression patterns could be studied in other biological samples (plasma, urine) in subjects with brain injury linked to aging, drug abuse, and stroke to identify reliable biomarkers that could be applied in clinical practice. Further studies with larger samples are needed to confirm these interesting findings.
Collapse
|
90
|
Histone deacetylase 3 inhibitors in learning and memory processes with special emphasis on benzamides. Eur J Med Chem 2019; 166:369-380. [DOI: 10.1016/j.ejmech.2019.01.077] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 12/24/2022]
|
91
|
Xu XF, Wang YC, Zong L, Wang XL. miR-151-5p modulates APH1a expression to participate in contextual fear memory formation. RNA Biol 2019; 16:282-294. [PMID: 30663934 DOI: 10.1080/15476286.2019.1572435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Long-term memory formation requires gene expression and new protein synthesis. MicroRNAs (miRNAs), a family of small non-coding RNAs that inhibit target gene mRNA expression, are involved in new memory formation. In this study, elevated miR-151-5p (miR-151) levels were found to be responsible for hippocampal contextual fear memory formation. Using a luciferase reporter assay, we demonstrated that miR-151 targets APH1a, a protein that has been identified as a key factor in γ-secretase activity, namely APH1a. Blocking miR-151 can upregulate APH1a protein levels and subsequently impair hippocampal fear memory formation. These results indicate that miR-151 is involved in hippocampal contextual fear memory by inhibiting APH1a protein expression. This work provides novel evidence for the role of miRNAs in memory formation and demonstrates the implication of APH1a protein in miRNA processing in the adult brain.
Collapse
Affiliation(s)
- Xu-Feng Xu
- a Institute of Brain Science and Disease, School of Basic Medicine , Qingdao University , Qingdao , Shandong , People's Republic of China.,b The Royal, Department of Psychiatry, and Department of Cellular and Molecular Medicine , University of Ottawa Institute of Mental Health Research , Ottawa , Canada.,c Department of Cell and Neurobiology , School of Basic Medicine, Shandong University , Jinan , Shandong , People's Republic of China
| | - You-Cui Wang
- a Institute of Brain Science and Disease, School of Basic Medicine , Qingdao University , Qingdao , Shandong , People's Republic of China
| | - Liang Zong
- d BGI-Shenzhen , Shenzhen , People's Republic of China
| | - Xiao-Long Wang
- e Department of Breast Surgery , Qilu hospital, Shandong University , Jinan , Shandong , People's Republic of China
| |
Collapse
|
92
|
Krauss S, Evert BO. The Role of MicroRNAs in Spinocerebellar Ataxia Type 3. J Mol Biol 2019; 431:1729-1742. [PMID: 30664869 DOI: 10.1016/j.jmb.2019.01.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/05/2019] [Accepted: 01/11/2019] [Indexed: 02/07/2023]
Abstract
More than 90% of the human genome are transcribed as non-coding RNAs. While it is still under debate if all these non-coding transcripts are functional, there is emerging evidence that RNA has several important functions in addition to coding for proteins. For example, microRNAs (miRNAs) are important regulatory RNAs that control gene expression in various biological processes and human diseases. In spinocerebellar ataxia type 3 (SCA3), a devastating neurodegenerative disease, miRNAs are involved in the disease process at different levels, including the deregulation of components of the general miRNA biogenesis machinery, as well as in the cell type-specific control of the expression of the SCA3 disease protein and other SCA3 disease-relevant proteins. However, it remains difficult to predict whether these changes are a cause or a consequence of the neurodegenerative process in SCA3. Further studies using standardized procedures for the analysis of miRNA expression and larger sample numbers are required to enhance our understanding of the miRNA-mediated processes involved in SCA3 disease and may enable the development of miRNA-based therapeutics. In this review, we summarize the findings of independent studies highlighting both the disease-related and cytoprotective roles of miRNAs that have been implicated so far in the disease process of SCA3.
Collapse
Affiliation(s)
- Sybille Krauss
- German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Street 27, 53127 Bonn, Germany
| | - Bernd O Evert
- Department of Neurology, University of Bonn, Sigmund-Freud-Street 25, 53127 Bonn, Germany.
| |
Collapse
|
93
|
Liu X, Jiao B, Shen L. The Epigenetics of Alzheimer's Disease: Factors and Therapeutic Implications. Front Genet 2018; 9:579. [PMID: 30555513 PMCID: PMC6283895 DOI: 10.3389/fgene.2018.00579] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 11/08/2018] [Indexed: 02/05/2023] Open
Abstract
Alzheimer’s disease (AD) is a well-known neurodegenerative disorder that imposes a great burden on the world. The mechanisms of AD are not yet fully understood. Current insight into the role of epigenetics in the mechanism of AD focuses on DNA methylation, remodeling of chromatin, histone modifications and non-coding RNA regulation. This review summarizes the current state of knowledge regarding the role of epigenetics in AD and the possibilities for epigenetically based therapeutics. The general conclusion is that epigenetic mechanisms play a variety of crucial roles in the development of AD, and there are a number of viable possibilities for treatments based on modulating these effects, but significant advances in knowledge and technology will be needed to move these treatments from the bench to the bedside.
Collapse
Affiliation(s)
- Xiaolei Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.,Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| |
Collapse
|
94
|
Shanesazzade Z, Peymani M, Ghaedi K, Nasr Esfahani MH. miR-34a/BCL-2 signaling axis contributes to apoptosis in MPP + -induced SH-SY5Y cells. Mol Genet Genomic Med 2018; 6:975-981. [PMID: 30221494 PMCID: PMC6305653 DOI: 10.1002/mgg3.469] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 07/26/2018] [Accepted: 08/09/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a neurodegenerative disorder which mainly affects the elderly population of various societies. The main hallmark of this disease is the loss of dopaminergic (DA) neurons. So far, numerous studies have implied the role of microRNAs in fine-tuning cellular processes including apoptosis. Studies have also shown that miR-34a is mainly involved in age-related disorders including Alzheimer's disease, and its expression is usually higher in the brain sample patients. Furthermore, the key role of miR-34a in the expression of BCL-2, and thus, in vitro and in vivo apoptosis has been revealed. miR-34a/BCL-2 axis is therefore of critical importance in inducing or inhibiting apoptosis. METHODS In this study, human SH-SY5Y cells were treated with MPP+ and the expression of miR-34a and BCL2 was assessed. RESULTS Our results also showed that treating human SH-SY5Y neuronal cells using MPP+ to induce oxidative stress and apoptosis led to the upregulation of miR-34a, as compared to the nontreated control group. Moreover, evaluating the expression level of BCL-2 in these cells indicated a contradictory pattern, as compared with miR-34a. It was also revealed that the expression of BCL-2 was significantly decreased in MPP+ -treated cells, thereby confirming previous studies regarding a new concept. In this study, we show that miR-34a/BCL-2 axis is directly correlated with oxidative stress and apoptosis in SH-SY5Y cells as a model of DA neurons. CONCLUSION miR-34a and its target gene, BCL-2, play a possible role in the induction of apoptosis in DA neurons, and therefore, they have a potential role in the pathogenesis of PD. Consequently, the therapeutic potential of miR-34a could be considered in order to inhibit the progression of PD.
Collapse
Affiliation(s)
- Zahra Shanesazzade
- Department of Biology, Faculty of Basic Sciences, Shahrekord BranchIslamic Azad UniversityShahrekordIran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord BranchIslamic Azad UniversityShahrekordIran
| | - Kamran Ghaedi
- Department of Biology, Faculty of SciencesUniversity of IsfahanIsfahanIran
- Department of Cellular Biotechnology, Cell Science Research CenterRoyan Institute for Biotechnology, ACECRIsfahanIran
| | | |
Collapse
|
95
|
Pardo M, Cheng Y, Sitbon YH, Lowell JA, Grieco SF, Worthen RJ, Desse S, Barreda-Diaz A. Insulin growth factor 2 (IGF2) as an emergent target in psychiatric and neurological disorders. Review. Neurosci Res 2018; 149:1-13. [PMID: 30389571 DOI: 10.1016/j.neures.2018.10.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/05/2018] [Accepted: 10/29/2018] [Indexed: 12/23/2022]
Abstract
Insulin-like growth factor 2 (IGF2) is abundantly expressed in the central nervous system (CNS). Recent evidence highlights the role of IGF2 in the brain, sustained by data showing its alterations as a common feature across a variety of psychiatric and neurological disorders. Previous studies emphasize the potential role of IGF2 in psychiatric and neurological conditions as well as in memory impairments, targeting IGF2 as a pro-cognitive agent. New research on animal models supports that upcoming investigations should explore IGF2's strong promising role as a memory enhancer. The lack of effective treatments for cognitive disturbances as a result of psychiatric diseases lead to further explore IGF2 as a promising target for the development of new pharmacology for the treatment of memory dysfunctions. In this review, we aim at gathering all recent relevant studies and findings on the role of IGF2 in the development of psychiatric diseases that occur with cognitive problems.
Collapse
Affiliation(s)
- M Pardo
- University of Miami Miller School of Medicine, Department of Neurology, Miami, FL, USA.
| | - Y Cheng
- University of California Los Angeles, Neurology Department, Los Angeles, CA, USA.
| | - Y H Sitbon
- University of Miami Miller School of Medicine, Department of Molecular and Cellular Pharmacology, Miami, FL, USA.
| | - J A Lowell
- University of Miami, Department of Psychiatry & Behavioral Sciences, Miami, FL, USA.
| | - S F Grieco
- University of California, Department of Anatomy and Neurobiology, Irvine, CA, USA.
| | - R J Worthen
- University of Miami, Department of Psychiatry & Behavioral Sciences, Miami, FL, USA.
| | - S Desse
- University of Miami, Department of Psychiatry & Behavioral Sciences, Miami, FL, USA.
| | - A Barreda-Diaz
- University of Miami Miller School of Medicine, Department of Neurology, Miami, FL, USA.
| |
Collapse
|
96
|
Fujita Y, Yamashita T. Sirtuins in Neuroendocrine Regulation and Neurological Diseases. Front Neurosci 2018; 12:778. [PMID: 30416425 PMCID: PMC6213750 DOI: 10.3389/fnins.2018.00778] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Silent information regulator 1 (SIRT1) is a mammalian homolog of the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin family. Sirtuin was originally studied as the lifespan-extending gene, silent information regulator 2 (SIRT2) in budding yeast. There are seven mammalian homologs of sirtuin (SIRT1–7), and SIRT1 is the closest homolog to SIRT2. SIRT1 modulates various key targets via deacetylation. In addition to histones, these targets include transcription factors, such as forkhead box O (FOXO), Ku70, p53, NF-κB, PPAR-gamma co-activator 1-alpha (PGC-1α), and peroxisome proliferator-activated receptor γ (PPARγ). SIRT1 has many biological functions, including aging, cell survival, differentiation, and metabolism. Genetic and physiological analyses in animal models have shown beneficial roles for SIRT1 in the brain during both development and adulthood. Evidence from in vivo and in vitro studies have revealed that SIRT1 regulates the cellular fate of neural progenitors, axon elongation, dendritic branching, synaptic plasticity, and endocrine function. In addition to its importance in physiological processes, SIRT1 has also been implicated in protection of neurons from degeneration in models of neurological diseases, such as traumatic brain injury and Alzheimer’s disease. In this review, we focus on the role of SIRT1 in the neuroendocrine system and neurodegenerative diseases. We also discuss the potential therapeutic implications of targeting the sirtuin pathway.
Collapse
Affiliation(s)
- Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan.,WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan.,WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.,Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
97
|
Fan SJ, Sun AB, Liu L. Epigenetic modulation during hippocampal development. Biomed Rep 2018; 9:463-473. [PMID: 30546873 DOI: 10.3892/br.2018.1160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/11/2018] [Indexed: 12/24/2022] Open
Abstract
The hippocampus is located in the limbic system and is vital in learning ability, memory formation and emotion regulation, and is associated with depression, epilepsy and mental retardation in an abnormal developmental situation. Several factors have been found to modulate the development of the hippocampus, and epigenetic modification have a crucial effect in this progress. The present review summarizes the epigenetic modifications, including DNA methylation, histone acetylation, and non-coding RNAs, regulating all stages of hippocampal development, focusing on the growth of Ammons horn and the dentate gyrus in humans and rodents. These modifications may significantly affect hippocampal development and health in addition to cognitive processes.
Collapse
Affiliation(s)
- Si-Jing Fan
- Department of Pharmacology, Medical School of Yangtze University, Jingzhou, Hubei 434023, P.R. China.,Laboratory of Neuronal and Brain Diseases Modulation, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - An-Bang Sun
- Laboratory of Neuronal and Brain Diseases Modulation, Yangtze University, Jingzhou, Hubei 434023, P.R. China.,Department of Anatomy, Medical School of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Lian Liu
- Department of Pharmacology, Medical School of Yangtze University, Jingzhou, Hubei 434023, P.R. China.,Laboratory of Neuronal and Brain Diseases Modulation, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| |
Collapse
|
98
|
Sierksma A, Lu A, Salta E, Vanden Eynden E, Callaerts-Vegh Z, D'Hooge R, Blum D, Buée L, Fiers M, De Strooper B. Deregulation of neuronal miRNAs induced by amyloid-β or TAU pathology. Mol Neurodegener 2018; 13:54. [PMID: 30314521 PMCID: PMC6186090 DOI: 10.1186/s13024-018-0285-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 09/27/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Despite diverging levels of amyloid-β (Aβ) and TAU pathology, different mouse models, as well as sporadic AD patients show predictable patterns of episodic memory loss. MicroRNA (miRNA) deregulation is well established in AD brain but it is unclear whether Aβ or TAU pathology drives those alterations and whether miRNA changes contribute to cognitive decline. METHODS miRNAseq was performed on cognitively intact (4 months) and impaired (10 months) male APPtg (APPswe/PS1L166P) and TAUtg (THY-Tau22) mice and their wild-type littermates (APPwt and TAUwt). We analyzed the hippocampi of 12 mice per experimental group (n = 96 in total), and employed a 2-way linear model to extract differentially expressed miRNAs. Results were confirmed by qPCR in a separate cohort of 4 M and 10 M APPtg and APPwt mice (n = 7-9 per group) and in human sporadic AD and non-demented control brain. Fluorescent in situ hybridization identified their cellular expression. Functional annotation of predicted targets was performed using GO enrichment. Behavior of wild-type mice was assessed after intracerebroventricular infusion of miRNA mimics. RESULTS Six miRNAs (miR-10a-5p, miR-142a-5p, miR-146a-5p, miR-155-5p, miR-211-5p, miR-455-5p) are commonly upregulated between APPtg and TAUtg mice, and four of these (miR-142a-5p, miR-146a-5p, miR-155-5p and miR-455-5p) are altered in AD patients. All 6 miRNAs are strongly enriched in neurons. Upregulating these miRNAs in wild-type mice is however not causing AD-related cognitive disturbances. CONCLUSION Diverging AD-related neuropathologies induce common disturbances in the expression of neuronal miRNAs. 4 of these miRNAs are also upregulated in AD patients. Therefore these 4 miRNAs (miR-142a-5p, miR-146a-5p, miR-155-5p and miR-455-5p) appear part of a core pathological process in AD patients and APPtg and TAUtg mice. They are however not causing cognitive disturbances in wild-type mice. As some of these miRNA target AD relevant proteins, they may be, in contrast, part of a protective response in AD.
Collapse
Affiliation(s)
- Annerieke Sierksma
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven research Institute for Neuroscience and Disease (LIND), KU Leuven, Leuven, Belgium
| | - Ashley Lu
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven research Institute for Neuroscience and Disease (LIND), KU Leuven, Leuven, Belgium
| | - Evgenia Salta
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven research Institute for Neuroscience and Disease (LIND), KU Leuven, Leuven, Belgium
| | - Elke Vanden Eynden
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven research Institute for Neuroscience and Disease (LIND), KU Leuven, Leuven, Belgium
| | - Zsuzsanna Callaerts-Vegh
- Faculty of Psychology and Educational Sciences, Laboratory of Biological Psychology, KU Leuven, Leuven, Belgium
| | - Rudi D'Hooge
- Faculty of Psychology and Educational Sciences, Laboratory of Biological Psychology, KU Leuven, Leuven, Belgium
| | - David Blum
- Université Lille, INSERM, CHU Lille, UMR-S 1172, LabEx DISTALZ, Alzheimer & Tauopathies, Lille, France
| | - Luc Buée
- Université Lille, INSERM, CHU Lille, UMR-S 1172, LabEx DISTALZ, Alzheimer & Tauopathies, Lille, France
| | - Mark Fiers
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- Department of Neurosciences, Leuven research Institute for Neuroscience and Disease (LIND), KU Leuven, Leuven, Belgium.
| | - Bart De Strooper
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- Department of Neurosciences, Leuven research Institute for Neuroscience and Disease (LIND), KU Leuven, Leuven, Belgium.
- Dementia Research Institute UK, ION, University College London, London, UK.
| |
Collapse
|
99
|
Abstract
Dementia is a complex clinical syndrome characterised by progressive decline in cognitive function. It usually presents itself as impairment in memory, loss of judgement, abstract thinking and other disturbances that are severe enough to interfere with activities of daily living. It has long been considered as one of the major challenges at present posing an ever-increasing demand on global health and social care systems. Of all the different forms of dementia, Alzheimer's disease (AD) is the most common. The term non-coding RNA (ncRNA) refers to RNA sequences which do not have the ability to be translated into proteins and therefore mainly fall within the realm of the recently acknowledged ‘dark matter’ of the genome. This genomic dark matter encompasses a whole spectrum of differing ncRNA families such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), PIWI-interacting RNAs (piRNAs), transfer RNAs (tRNAs), small nuclear RNAs (snoRNAs) and circular RNAs (circRNAs), to name but a few. Consequently, due to the widespread influences of miRNAs and lncRNAs across all disease pathways, it is of critical importance for researchers in the field of dementia to focus their attention on possible ncRNA-induced pathogeneses, with the ultimate goal of identifying novel diagnostic procedures and drug targets, together with the development of novel therapies to control such a devastating mental condition in the patient population.
Collapse
|
100
|
Higuchi Y, Soga T, Parhar IS. Potential Roles of microRNAs in the Regulation of Monoamine Oxidase A in the Brain. Front Mol Neurosci 2018; 11:339. [PMID: 30271325 PMCID: PMC6149293 DOI: 10.3389/fnmol.2018.00339] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/28/2018] [Indexed: 12/15/2022] Open
Abstract
Monoamine oxidase A (MAO-A) is an enzyme that regulates the levels of monoamine neurotransmitters, such as serotonin, noradrenaline and dopamine and it has been used as a therapeutic target for depression. However, MAO-A inhibitors, which directly acts on MAO-A protein, have limited use due to their adverse effects. microRNAs (miRNAs) are 18-22 nucleotide long, small non-coding RNAs, which have recently emerged as regulators of protein levels that could potentially be new therapeutic targets for psychiatric disorders. This review article aims to discuss the current status of the treatment for depression with MAO-A inhibitors and the regulatory factors of MAO-A. Further, the review also proposes possible regulatory mechanisms of MAO-A by miRNAs, which leads to better understanding of the pathology of depressive disorders and their potential use as therapeutic agents.
Collapse
Affiliation(s)
| | | | - Ishwar S. Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|