101
|
Kang Q, Xiang Y, Li D, Liang J, Zhang X, Zhou F, Qiao M, Nie Y, He Y, Cheng J, Dai Y, Li Y. MiR-124-3p attenuates hyperphosphorylation of Tau protein-induced apoptosis via caveolin-1-PI3K/Akt/GSK3β pathway in N2a/APP695swe cells. Oncotarget 2018; 8:24314-24326. [PMID: 28186985 PMCID: PMC5421849 DOI: 10.18632/oncotarget.15149] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/24/2017] [Indexed: 01/01/2023] Open
Abstract
Hyperphosphorylation of Tau forming neurofibrillary tangles has been considered as a crucial event in the pathogenesis of Alzheimer's disease (AD). MiR-124-3p belongs to microRNA (miRNA) family and was markedly decreased in AD, however, the functions of miR-124-3p in the pathogenesis of AD remain unknown. We observed that the expression of miR-124-3p was significantly decreased in N2a/APP695swe cells; and transfection of miR-124-3p mimics not only attenuated cell apoptosis and abnormal hyperphosphorylation of Tau protein without any changes of total Tau protein, but also increased expression levels of Caveolin-1, phosphoinositide 3-kinase (PI3K), phospho-Akt (Akt-Ser473)/Akt, phospho-glycogen synthase kinase-3 beta (GSK-3β-Ser9)/GSK-3β in N2a/APP695swe cells. We further found that miR-12-3p directly targeted Caveolin-1; miR-124-3p inhibited abnormal hyperphosphorylation of Tau by regulating Caveolin-1-PI3K/Akt/GSK3β pathway in AD. This study reveals that miR-124-3p may play a neuroprotective role in AD, which may provide new ideas and therapeutic targets for AD.
Collapse
Affiliation(s)
- Qingmei Kang
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, China.,Center for Molecular Medicine Testing, Chongqing Medical University, Chongqing, 400016, China
| | - Yue Xiang
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, China.,Center for Molecular Medicine Testing, Chongqing Medical University, Chongqing, 400016, China
| | - Dan Li
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, China.,Center for Molecular Medicine Testing, Chongqing Medical University, Chongqing, 400016, China
| | - Jie Liang
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, China.,Center for Molecular Medicine Testing, Chongqing Medical University, Chongqing, 400016, China
| | - Xiong Zhang
- Center for Molecular Medicine Testing, Chongqing Medical University, Chongqing, 400016, China
| | - Fanlin Zhou
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, China.,Center for Molecular Medicine Testing, Chongqing Medical University, Chongqing, 400016, China
| | - Mengyuan Qiao
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, China.,Center for Molecular Medicine Testing, Chongqing Medical University, Chongqing, 400016, China
| | - Yingling Nie
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, China.,Center for Molecular Medicine Testing, Chongqing Medical University, Chongqing, 400016, China
| | - Yurong He
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, China.,Center for Molecular Medicine Testing, Chongqing Medical University, Chongqing, 400016, China
| | - Jingyi Cheng
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, China.,Center for Molecular Medicine Testing, Chongqing Medical University, Chongqing, 400016, China
| | - Yubing Dai
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yu Li
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, China.,Center for Molecular Medicine Testing, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
102
|
Miquel S, Champ C, Day J, Aarts E, Bahr BA, Bakker M, Bánáti D, Calabrese V, Cederholm T, Cryan J, Dye L, Farrimond JA, Korosi A, Layé S, Maudsley S, Milenkovic D, Mohajeri MH, Sijben J, Solomon A, Spencer JPE, Thuret S, Vanden Berghe W, Vauzour D, Vellas B, Wesnes K, Willatts P, Wittenberg R, Geurts L. Poor cognitive ageing: Vulnerabilities, mechanisms and the impact of nutritional interventions. Ageing Res Rev 2018; 42:40-55. [PMID: 29248758 DOI: 10.1016/j.arr.2017.12.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/08/2017] [Accepted: 12/08/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Ageing is a highly complex process marked by a temporal cascade of events, which promote alterations in the normal functioning of an individual organism. The triggers of normal brain ageing are not well understood, even less so the factors which initiate and steer the neuronal degeneration, which underpin disorders such as dementia. A wealth of data on how nutrients and diets may support cognitive function and preserve brain health are available, yet the molecular mechanisms underlying their biological action in both normal ageing, age-related cognitive decline, and in the development of neurodegenerative disorders have not been clearly elucidated. OBJECTIVES This review aims to summarise the current state of knowledge of vulnerabilities that predispose towards dysfunctional brain ageing, highlight potential protective mechanisms, and discuss dietary interventions that may be used as therapies. A special focus of this paper is on the impact of nutrition on neuroprotection and the underlying molecular mechanisms, and this focus reflects the discussions held during the 2nd workshop 'Nutrition for the Ageing Brain: Functional Aspects and Mechanisms' in Copenhagen in June 2016. The present review is the most recent in a series produced by the Nutrition and Mental Performance Task Force under the auspice of the International Life Sciences Institute Europe (ILSI Europe). CONCLUSION Coupling studies of cognitive ageing with studies investigating the effect of nutrition and dietary interventions as strategies targeting specific mechanisms, such as neurogenesis, protein clearance, inflammation, and non-coding and microRNAs is of high value. Future research on the impact of nutrition on cognitive ageing will need to adopt a longitudinal approach and multimodal nutritional interventions will likely need to be imposed in early-life to observe significant impact in older age.
Collapse
Affiliation(s)
- Sophie Miquel
- Mars-Wrigley, 1132 W. Blackhawk Street, Chicago, IL 60642, United States
| | - Claire Champ
- Human Appetite Research Unit, School of Psychology, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Jon Day
- Cerebrus Associates Limited, The White House, 2 Meadrow, Godalming, Surrey, GU7 3HN, United Kingdom
| | - Esther Aarts
- Donders Institute for Brain, Cognition, and Behaviour, Radboud University Nijmegen, Kapittelweg 29, 6525 EN Nijmegen, The Netherlands
| | - Ben A Bahr
- Biotechnology Research and Training Centre, University of North Carolina - Pembroke, United States
| | - Martijntje Bakker
- The Netherlands Organisation for Health Research and Development, Laan van Nieuw Oost-Indië 334, 2593 CE The Hague, The Netherlands
| | - Diána Bánáti
- International Life Sciences Institute, Europe (ILSI Europe), Av E. Mounier 83, Box 6, 1200 Brussels, Belgium
| | - Vittorio Calabrese
- University of Catania, Department of Biomedical and Biotechnological Sciences, Biological Tower - Via Santa Sofia, 97, Catania, Italy
| | - Tommy Cederholm
- University of Uppsala, Institutionen för folkhälso- och vårdvetenskap, Klinisk nutrition och metabolism, Uppsala Science Park, 751 85 Uppsala, Sweden
| | - John Cryan
- Anatomy & Neuroscience, University College Cork, 386 Western Gateway Building, Cork, Ireland
| | - Louise Dye
- Human Appetite Research Unit, School of Psychology, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | | | - Aniko Korosi
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Sophie Layé
- Nutrition et Neurobiologie Intégrée, INRA Bordeaux University, 146 rue Léo Saignat, 33076 Bordeaux cedex, France
| | - Stuart Maudsley
- Department of Biomedical Research and VIB-UAntwerp Center for Molecular Neurology, University of Antwerp, Gebouw V, Campus Drie Eiken, Universiteitsplein 1, 2610 Antwerpen, Belgium
| | - Dragan Milenkovic
- INRA, Human Nutrition Unit, UCA, F-63003, Clermont-Ferrand, France; Department of Internal Medicine, Division of Cardiovascular Medicine, School of Medicine, University of California Davis, Davis, CA 95616, United States
| | - M Hasan Mohajeri
- DSM Nutritional Products Ltd., Wurmisweg 576, Kaiseraugst 4303, Switzerland
| | - John Sijben
- Nutricia Research, Nutricia Advanced Medical Nutrition, PO Box 80141, 3508TC, Utrecht, The Netherlands
| | - Alina Solomon
- Aging Research Center, Karolinska Institutet, Gävlegatan 16, SE-113 30 Stockholm, Sweden
| | - Jeremy P E Spencer
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading, RG6 6AP, United Kingdom
| | - Sandrine Thuret
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, The Maurice Wohl Clinical Neuroscience Institute,125 Coldharbour Lane, SE5 9NU London, United Kingdom
| | - Wim Vanden Berghe
- PPES, Department Biomedical Sciences, University Antwerp, Campus Drie Eiken, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - David Vauzour
- University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - Bruno Vellas
- Department of Geriatric Medicine, CHU Toulouse, Gerontopole, Toulouse, France
| | - Keith Wesnes
- Wesnes Cognition Limited, Little Paddock, Streatley on Thames, RG8 9RD, United Kingdom; Medical School, University of Exeter, Exeter, United Kingdom; Department of Psychology, Northumbria University, Newcastle, United Kingdom; Centre for Human Psychopharmacology, Swinburne University, Melbourne, Australia; Medicinal Plant Research Group, Newcastle University, United Kingdom
| | - Peter Willatts
- School of Psychology, University of Dundee Nethergate, Dundee, DD1 4HN, United Kingdom
| | - Raphael Wittenberg
- London School of Economics and Political Science, Personal Social Services Research Unit, London, United Kingdom
| | - Lucie Geurts
- International Life Sciences Institute, Europe (ILSI Europe), Av E. Mounier 83, Box 6, 1200 Brussels, Belgium.
| |
Collapse
|
103
|
Idda ML, Munk R, Abdelmohsen K, Gorospe M. Noncoding RNAs in Alzheimer's disease. WILEY INTERDISCIPLINARY REVIEWS. RNA 2018; 9. [PMID: 29327503 PMCID: PMC5847280 DOI: 10.1002/wrna.1463] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/24/2017] [Accepted: 11/29/2017] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the main cause of dementia among the elderly worldwide. Despite intense efforts to develop drugs for preventing and treating AD, no effective therapies are available as yet, posing a growing burden at the personal, medical, and socioeconomic levels. AD is characterized by the production and aggregation of amyloid β (Aβ) peptides derived from amyloid precursor protein (APP), the presence of hyperphosphorylated microtubule-associated protein Tau (MAPT), and chronic inflammation leading to neuronal loss. Aβ accumulation and hyperphosphorylated Tau are responsible for the main histopathological features of AD, Aβ plaques, and neurofibrillary tangles (NFTs), respectively. However, the full spectrum of molecular factors that contribute to AD pathogenesis is not known. Noncoding (nc)RNAs, including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), regulate gene expression at the transcriptional and posttranscriptional levels in various diseases, serving as biomarkers and potential therapeutic targets. There is rising recognition that ncRNAs have been implicated in both the onset and pathogenesis of AD. Here, we review the ncRNAs implicated posttranscriptionally in the main AD pathways and discuss the growing interest in targeting regulatory ncRNAs therapeutically to combat AD pathology. WIREs RNA 2018, 9:e1463. doi: 10.1002/wrna.1463 This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- M Laura Idda
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland
| | - Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
104
|
Abstract
Significant advancements have been made in unraveling and understanding the non-coding elements of the human genome. New insights into the structure and function of noncoding RNAs have emerged. Their relevance in the context of both physiological cellular homeostasis and human diseases is getting appreciated. As a result, exploration of noncoding RNAs, in particular microRNAs (miRs), as therapeutic agents or targets of therapeutic strategies is under way. This review summarizes and discusses in depth the current literature on the role of miRs in neurodegenerative diseases.
Collapse
Affiliation(s)
- Salil Sharma
- Department of Psychological and Brain Sciences, The Linda and Jack Gill Center for Bimolecular Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Hui-Chen Lu
- Department of Psychological and Brain Sciences, The Linda and Jack Gill Center for Bimolecular Sciences, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
105
|
Burak K, Lamoureux L, Boese A, Majer A, Saba R, Niu Y, Frost K, Booth SA. MicroRNA-16 targets mRNA involved in neurite extension and branching in hippocampal neurons during presymptomatic prion disease. Neurobiol Dis 2017; 112:1-13. [PMID: 29277556 DOI: 10.1016/j.nbd.2017.12.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 11/14/2017] [Accepted: 12/19/2017] [Indexed: 12/21/2022] Open
Abstract
The mechanisms that lead to neuronal death in neurodegenerative diseases are poorly understood. Prion diseases, like many more common disorders such as Alzheimer's and Parkinson's diseases, are characterized by the progressive accumulation of misfolded disease-specific proteins. The earliest changes observed in brain tissue include a reduction in synaptic number and retraction of dendritic spines, followed by reduced length and branching of neurites. These pathologies are observable during presymptomatic stages of disease and are accompanied by altered expression of transcripts that include miRNAs. Here we report that miR-16 localized within hippocampal CA1 neurons is increased during early prion disease. Modulating miR-16 expression in mature murine hippocampal neurons by expression from a lentivirus, thus mimicking the modest increase seen in vivo, was found to induce neurodegeneration. This was characterized by retraction of neurites and reduced branching. We performed immunoprecipitation of the miR-16 enriched RISC complex, and identified associated transcripts from the co-immunoprecipitated RNA (Ago2 RIP-Chip). These transcripts were enriched with predicted binding sites for miR-16, including the validated miR-16 targets APP and BCL2, as well as numerous novel targets. In particular, genes within the neurotrophin receptor mediated MAPK/ERK pathway were potentially regulated by miR-16; including TrkB (NTRK2), MEK1 (MAP2K1) and c-Raf (RAF). Increased miR-16 expression in neurons during presymptomatic prion disease and reduction in proteins involved in MAPK/ERK signaling represents a possible mechanism by which neurite length and branching are decreased during early stages of disease.
Collapse
Affiliation(s)
- Kristyn Burak
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada; Department of Medical Microbiology and Infectious Diseases, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lise Lamoureux
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Amrit Boese
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada; Department of Medical Microbiology and Infectious Diseases, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Anna Majer
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada; Department of Medical Microbiology and Infectious Diseases, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Reuben Saba
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Yulian Niu
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Kathy Frost
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Stephanie A Booth
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada; Department of Medical Microbiology and Infectious Diseases, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
106
|
Lu J, Xu Y, Quan Z, Chen Z, Sun Z, Qing H. Dysregulated microRNAs in neural system: Implication in pathogenesis and biomarker development in Parkinson’s disease. Neuroscience 2017; 365:70-82. [DOI: 10.1016/j.neuroscience.2017.09.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/14/2017] [Accepted: 09/15/2017] [Indexed: 01/03/2023]
|
107
|
Identification of genome-wide non-canonical spliced regions and analysis of biological functions for spliced sequences using Read-Split-Fly. BMC Bioinformatics 2017; 18:382. [PMID: 28984182 PMCID: PMC5629565 DOI: 10.1186/s12859-017-1801-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background It is generally thought that most canonical or non-canonical splicing events involving U2- and U12 spliceosomes occur within nuclear pre-mRNAs. However, the question of whether at least some U12-type splicing occurs in the cytoplasm is still unclear. In recent years next-generation sequencing technologies have revolutionized the field. The “Read-Split-Walk” (RSW) and “Read-Split-Run” (RSR) methods were developed to identify genome-wide non-canonical spliced regions including special events occurring in cytoplasm. As the significant amount of genome/transcriptome data such as, Encyclopedia of DNA Elements (ENCODE) project, have been generated, we have advanced a newer more memory-efficient version of the algorithm, “Read-Split-Fly” (RSF), which can detect non-canonical spliced regions with higher sensitivity and improved speed. The RSF algorithm also outputs the spliced sequences for further downstream biological function analysis. Results We used open access ENCODE project RNA-Seq data to search spliced intron sequences against the U12-type spliced intron sequence database to examine whether some events could occur as potential signatures of U12-type splicing. The check was performed by searching spliced sequences against 5’ss and 3’ss sequences from the well-known orthologous U12-type spliceosomal intron database U12DB. Preliminary results of searching 70 ENCODE samples indicated that the presence of 5’ss with U12-type signature is more frequent than U2-type and prevalent in non-canonical junctions reported by RSF. The selected spliced sequences have also been further studied using miRBase to elucidate their functionality. Preliminary results from 70 samples of ENCODE datasets show that several miRNAs are prevalent in studied ENCODE samples. Two of these are associated with many diseases as suggested in the literature. Specifically, hsa-miR-1273 and hsa-miR-548 are associated with many diseases and cancers. Conclusions Our RSF pipeline is able to detect many possible junctions (especially those with a high RPKM) with very high overall accuracy and relative high accuracy for novel junctions. We have incorporated useful parameter features into the pipeline such as, handling variable-length read data, and searching spliced sequences for splicing signatures and miRNA events. We suggest RSF, a tool for identifying novel splicing events, is applicable to study a range of diseases across biological systems under different experimental conditions. Electronic supplementary material The online version of this article (10.1186/s12859-017-1801-y) contains supplementary material, which is available to authorized users.
Collapse
|
108
|
Resveratrol as a Natural Autophagy Regulator for Prevention and Treatment of Alzheimer's Disease. Nutrients 2017; 9:nu9090927. [PMID: 28837083 PMCID: PMC5622687 DOI: 10.3390/nu9090927] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/03/2017] [Accepted: 08/11/2017] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most common neurodegenerative disorders over the age of 65 years old. Although several underlying mechanisms for explaining the pathogenesis of AD are elucidated, the effective supplements or drugs for the intervention of AD are still limited. Recently, impaired autophagy associated with miRNA dysfunction has been reported to involve in aging and aging-related neurodegenerative diseases. Thus, the activation of autophagy through effectively regulating miRNAs may become a potential target for the prevention or treatment of AD. Mounting evidence from in vitro and in vivo AD models has demonstrated that resveratrol, one of polyphenolic compounds, can exert neuroprotective role in neurodegenerative diseases especially AD. In this review, the regulation of miRNAs and autophagy using resveratrol during the prevention and treatment of AD are systematically discussed, which will be beneficial to establish a target for the direct link between pharmacological intervention and AD in the future.
Collapse
|
109
|
Abstract
The emerging complexity of the transcriptional landscape poses great challenges to our conventional preconceptions of how the genome regulates brain function and dysfunction. Non-protein-coding RNAs (ncRNAs) confer a high level of intricate and dynamic regulation of various molecular processes in the CNS and they have been implicated in neurodevelopment and brain ageing, as well as in synapse function and cognitive performance, in both health and disease. ncRNA-mediated processes may be involved in various aspects of the pathogenesis of neurodegenerative disorders. Understanding these events may help to develop novel diagnostic and therapeutic tools. Here, we provide an overview of the complex mechanisms that are affected by the diverse ncRNA classes that have been implicated in neurodegeneration.
Collapse
|
110
|
Kovalchuk A, Ilnytskyy Y, Rodriguez-Juarez R, Katz A, Sidransky D, Kolb B, Kovalchuk O. Growth of malignant extracranial tumors alters microRNAome in the prefrontal cortex of TumorGraft mice. Oncotarget 2017; 8:88276-88293. [PMID: 29179434 PMCID: PMC5687604 DOI: 10.18632/oncotarget.19835] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 04/27/2017] [Indexed: 12/13/2022] Open
Abstract
A wide array of central nervous system complications, neurological deficits, and cognitive impairments occur and persist as a result of systemic cancer and cancer treatments. This condition is known as chemo brain and it affects over half of cancer survivors. Recent studies reported that cognitive impairments manifest before chemotherapy and are much broader than chemo brain alone, thereby adding in tumor brain as a component. The molecular mechanisms of chemo brain are under-investigated, and the mechanisms of tumor brain have not been analyzed at all. The frequency and timing, as well as the long-term persistence, of chemo brain and tumor brain suggest they may be epigenetic in nature. MicroRNAs, small, single-stranded non-coding RNAs, constitute an important part of the cellular epigenome and are potent regulators of gene expression. miRNAs are crucial for brain development and function, and are affected by a variety of different stresses, diseases and conditions. However, nothing is known about the effects of extracranial tumor growth or chemotherapy agents on the brain microRNAome. We used the well-established TumorGraft ™ mouse models of triple negative (TNBC) and progesterone receptor positive (PR+BC) breast cancer, and profiled global microRNAome changes in tumor-bearing mice upon chemotherapy, as compared to untreated tumor-bearing mice and intact mice. Our analysis focused on the prefrontal cortex (PFC), based on its roles in memory, learning, and executive functions, and on published data showing the PFC is a target in chemo brain. This is the first study showing that tumor presence alone significantly impacted the small RNAome of PFC tissues. Both tumor growth and chemotherapy treatment affected the small RNAome and altered levels of miRNAs, piRNAs, tRNAs, tRNA fragments and other molecules involved in post-transcriptional regulation of gene expression. Amongst those, miRNA changes were the most pronounced, involving several miRNA families, such as the miR-200 family and miR-183/96/182 cluster; both were deregulated in tumor-bearing and chemotherapy-treated animals. We saw that miRNA deregulation was associated with altered levels of brain-derived neurotrophic factor (BDNF), which plays an important role in cognition and memory and is one of the known miRNA targets. BDNF downregulation has been associated with an array of neurological conditions and could be one of the mechanisms underlying tumor brain and chemo brain. In the future our study could serve as a roadmap for further analysis of cancer and chemotherapy's neural side effects, and differentially expressed miRNAs should be explored as potential tumor brain and chemo brain biomarkers.
Collapse
Affiliation(s)
- Anna Kovalchuk
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada.,Leaders in Medicine Program, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Yaroslav Ilnytskyy
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB, Canada
| | | | - Amanda Katz
- Department of Oncology, Champions Oncology, Baltimore, MD, USA
| | - David Sidransky
- Department of Oncology, Champions Oncology, Baltimore, MD, USA
| | - Bryan Kolb
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB, Canada
| |
Collapse
|
111
|
Gaudet AD, Fonken LK, Watkins LR, Nelson RJ, Popovich PG. MicroRNAs: Roles in Regulating Neuroinflammation. Neuroscientist 2017; 24:221-245. [PMID: 28737113 DOI: 10.1177/1073858417721150] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that broadly affect cellular and physiological function in all multicellular organisms. Here, the role of miRNAs in neuroinflammation is considered. miRNAs are 21- to 23-oligonucleotide RNAs that regulate translation of specific RNAs by binding to complementary regulatory RNA sequences, thereby causing mRNA degradation or sequestration. More than 5000 miRNAs likely exist in humans, and each miRNA binds an average of 200 RNAs. Specific immunomodulatory miRNAs can regulate a set of RNAs in a coordinated manner, suggesting that effective miRNA-based therapeutic manipulations for neuroinflammatory conditions may be revealed. For instance, miRNAs that preferentially inhibit translation of many cellular anti-inflammatory proteins could drive a pro-inflammatory response. Key pro-inflammatory ( miR-155, miR-27b, miR-326), anti-inflammatory ( miR-124, miR-146a, miR-21, miR-223), and mixed immunomodulatory ( let-7 family) miRNAs regulate neuroinflammation in various pathologies, including spinal cord injury, multiple sclerosis, ischemic stroke, and Alzheimer's disease. miRNAs represent a newly revealed layer of physiological complexity, the therapeutic benefits of which remain to be fully explored and exploited. In this review, we discuss the role of miRNAs in neuroinflammatory regulation and discuss how controlling miRNAs could alter cellular machinery to improve neuroinflammatory dynamics.
Collapse
Affiliation(s)
- Andrew D Gaudet
- 1 Center for Neuroscience, University of Colorado Boulder, CO, USA.,2 Department of Psychology and Neuroscience, University of Colorado Boulder, CO, USA
| | - Laura K Fonken
- 1 Center for Neuroscience, University of Colorado Boulder, CO, USA.,2 Department of Psychology and Neuroscience, University of Colorado Boulder, CO, USA
| | - Linda R Watkins
- 1 Center for Neuroscience, University of Colorado Boulder, CO, USA.,2 Department of Psychology and Neuroscience, University of Colorado Boulder, CO, USA
| | - Randy J Nelson
- 3 Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.,4 Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Phillip G Popovich
- 3 Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.,4 Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.,5 Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
112
|
Zhao J, Yue D, Zhou Y, Jia L, Wang H, Guo M, Xu H, Chen C, Zhang J, Xu L. The Role of MicroRNAs in Aβ Deposition and Tau Phosphorylation in Alzheimer's Disease. Front Neurol 2017; 8:342. [PMID: 28769871 PMCID: PMC5513952 DOI: 10.3389/fneur.2017.00342] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 06/30/2017] [Indexed: 12/22/2022] Open
Abstract
Alzheimer’s disease (AD), with main clinical features of progressive impairment in cognitive and behavioral functions, is the most common degenerative disease of the central nervous system. Recent evidence showed that microRNAs (miRNAs) played important roles in the pathological progression of AD. In this article, we reviewed the promising role of miRNAs in both Aβ deposition and Tau phosphorylation, two key pathological characters in the pathological progression of AD, which might be helpful for the understanding of pathogenesis and the development of new strategies of clinical diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Juanjuan Zhao
- Department of Immunology, Zunyi Medical College, Guizhou, China
| | - Dongxu Yue
- Department of Immunology, Zunyi Medical College, Guizhou, China
| | - Ya Zhou
- Department of Medical Physics, Zunyi Medical College, Guizhou, China
| | - Li Jia
- Department of Immunology, Zunyi Medical College, Guizhou, China
| | - Hairong Wang
- Department of Immunology, Zunyi Medical College, Guizhou, China
| | - Mengmeng Guo
- Department of Immunology, Zunyi Medical College, Guizhou, China
| | - Hualin Xu
- Department of Immunology, Zunyi Medical College, Guizhou, China
| | - Chao Chen
- Department of Immunology, Zunyi Medical College, Guizhou, China
| | - Jidong Zhang
- Department of Immunology, Zunyi Medical College, Guizhou, China
| | - Lin Xu
- Department of Immunology, Zunyi Medical College, Guizhou, China
| |
Collapse
|
113
|
Chen JJ, Zhao B, Zhao J, Li S. Potential Roles of Exosomal MicroRNAs as Diagnostic Biomarkers and Therapeutic Application in Alzheimer's Disease. Neural Plast 2017; 2017:7027380. [PMID: 28770113 PMCID: PMC5523215 DOI: 10.1155/2017/7027380] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 05/22/2017] [Indexed: 12/21/2022] Open
Abstract
Exosomes are bilipid layer-enclosed vesicles derived from endosomes and are released from neural cells. They contain a diversity of proteins, mRNAs, and microRNAs (miRNAs) that are delivered to neighboring cells and/or are transported to distant sites. miRNAs released from exosomes appear to be associated with multiple neurodegenerative conditions linking to Alzheimer's disease (AD) which is marked by hyperphosphorylated tau proteins and accumulation of Aβ plaques. Exciting findings reveal that miRNAs released from exosomes modulate the expression and function of amyloid precursor proteins (APP) and tau proteins. These open up the possibility that dysfunctional exosomal miRNAs may influence AD progression. In addition, it has been confirmed that the interaction between miRNAs released by exosomes and Toll-like receptors (TLR) initiates inflammation. In exosome support-deprived neurons, exosomal miRNAs may regulate neuroplasticity to relieve neurological damage. In this review, we summarize the literature on the function of exosomal miRNAs in AD pathology, the potential of these miRNAs as diagnostic biomarkers in AD, and the use of exosomes in the delivery of miRNAs which may lead to major advances in the field of macromolecular drug delivery.
Collapse
Affiliation(s)
- Jian-jiao Chen
- Department of Physiology, Liaoning Provincial Key Laboratory of Cerebral Diseases, Dalian Medical University, Dalian City, Liaoning Province 116044, China
- Department of General Surgery, Subei People's Hospital of Jiangsu Province, Yangzhou City, Jiangsu Province 225000, China
| | - Bin Zhao
- Technology Centre of Target-Based Nature Products for Prevention and Treatment of Aging-Related Neurodegeneration, Dalian Medical University, Dalian, Liaoning, China
| | - Jie Zhao
- Department of Physiology, Liaoning Provincial Key Laboratory of Cerebral Diseases, Dalian Medical University, Dalian City, Liaoning Province 116044, China
- Technology Centre of Target-Based Nature Products for Prevention and Treatment of Aging-Related Neurodegeneration, Dalian Medical University, Dalian, Liaoning, China
| | - Shao Li
- Department of Physiology, Liaoning Provincial Key Laboratory of Cerebral Diseases, Dalian Medical University, Dalian City, Liaoning Province 116044, China
| |
Collapse
|
114
|
Yagishita S, Hirasawa A. Intermittent hypoxia produces Alzheimer disease? Oncotarget 2017; 8:41786-41787. [PMID: 28562318 PMCID: PMC5522025 DOI: 10.18632/oncotarget.18214] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 05/24/2017] [Indexed: 01/26/2023] Open
Affiliation(s)
- Sosuke Yagishita
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Akira Hirasawa
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
115
|
Derghal A, Djelloul M, Trouslard J, Mounien L. The Role of MicroRNA in the Modulation of the Melanocortinergic System. Front Neurosci 2017; 11:181. [PMID: 28424580 PMCID: PMC5380727 DOI: 10.3389/fnins.2017.00181] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 03/20/2017] [Indexed: 11/13/2022] Open
Abstract
The central control of energy balance involves a highly regulated neuronal network within the hypothalamus and the dorsal vagal complex. In these structures, pro-opiomelanocortin (POMC) neurons are known to reduce meal size and to increase energy expenditure. In addition, leptin, a peripheral signal that relays information regarding body fat content, modulates the activity of melanocortin pathway neurons including POMC-, Agouti-related peptide (AgRP)/Neuropeptide Y (NPY)-, melanocortin receptors (MC3R and MC4R)-expressing neurons. MicroRNAs (miRNAs) are short non-coding RNAs of 22–26 nucleotides that post-transcriptionally interfere with target gene expression by binding to their mRNAs. Evidence has demonstrated that miRNAs play important roles in the central regulation of energy balance. In this context, different studies identified miRNAs including miR-200 family, miR-103, or miR-488 that could target the genes of melanocortin pathway. More precisely, these different miRNAs can modulate energy homeostasis by affecting leptin transduction pathway in the POMC, or AgRP/NPY neurons. This article reviews the role of identified miRNAs in the modulation of melanocortin pathway in the context of energy homeostasis.
Collapse
Affiliation(s)
- Adel Derghal
- Physiologie et Physiopathologie du Système Nerveux Somatomoteur et Neurovégétatif (PPSN), Aix Marseille UniversityMarseille, France
| | - Mehdi Djelloul
- Physiologie et Physiopathologie du Système Nerveux Somatomoteur et Neurovégétatif (PPSN), Aix Marseille UniversityMarseille, France.,Department of Cell and Molecular Biology, Karolinska InstituteStockholm, Sweden
| | - Jérôme Trouslard
- Physiologie et Physiopathologie du Système Nerveux Somatomoteur et Neurovégétatif (PPSN), Aix Marseille UniversityMarseille, France
| | - Lourdes Mounien
- Physiologie et Physiopathologie du Système Nerveux Somatomoteur et Neurovégétatif (PPSN), Aix Marseille UniversityMarseille, France
| |
Collapse
|
116
|
Millan MJ. Linking deregulation of non-coding RNA to the core pathophysiology of Alzheimer's disease: An integrative review. Prog Neurobiol 2017; 156:1-68. [PMID: 28322921 DOI: 10.1016/j.pneurobio.2017.03.004] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/09/2017] [Accepted: 03/09/2017] [Indexed: 02/06/2023]
Abstract
The human genome encodes a vast repertoire of protein non-coding RNAs (ncRNA), some specific to the brain. MicroRNAs, which interfere with the translation of target mRNAs, are of particular interest since their deregulation has been implicated in neurodegenerative disorders like Alzheimer's disease (AD). However, it remains challenging to link the complex body of observations on miRNAs and AD into a coherent framework. Using extensive graphical support, this article discusses how a diverse panoply of miRNAs convergently and divergently impact (and are impacted by) core pathophysiological processes underlying AD: neuroinflammation and oxidative stress; aberrant generation of β-amyloid-42 (Aβ42); anomalies in the production, cleavage and post-translational marking of Tau; impaired clearance of Aβ42 and Tau; perturbation of axonal organisation; disruption of synaptic plasticity; endoplasmic reticulum stress and the unfolded protein response; mitochondrial dysfunction; aberrant induction of cell cycle re-entry; and apoptotic loss of neurons. Intriguingly, some classes of miRNA provoke these cellular anomalies, whereas others act in a counter-regulatory, protective mode. Moreover, changes in levels of certain species of miRNA are a consequence of the above-mentioned anomalies. In addition to miRNAs, circular RNAs, piRNAs, long non-coding RNAs and other types of ncRNA are being increasingly implicated in AD. Overall, a complex mesh of deregulated and multi-tasking ncRNAs reciprocally interacts with core pathophysiological mechanisms underlying AD. Alterations in ncRNAs can be detected in CSF and the circulation as well as the brain and are showing promise as biomarkers, with the ultimate goal clinical exploitation as targets for novel modes of symptomatic and course-altering therapy.
Collapse
Affiliation(s)
- Mark J Millan
- Centre for Therapeutic Innovation in Neuropsychiatry, institut de recherche Servier, 125 chemin de ronde, 78290 Croissy sur Seine, France.
| |
Collapse
|
117
|
Reddy PH, Tonk S, Kumar S, Vijayan M, Kandimalla R, Kuruva CS, Reddy AP. A critical evaluation of neuroprotective and neurodegenerative MicroRNAs in Alzheimer's disease. Biochem Biophys Res Commun 2017; 483:1156-1165. [PMID: 27524239 PMCID: PMC5343756 DOI: 10.1016/j.bbrc.2016.08.067] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 08/10/2016] [Indexed: 12/31/2022]
Abstract
Currently, 5.4 million Americans suffer from AD, and these numbers are expected to increase up to 16 million by 2050. Despite tremendous research efforts, we still do not have drugs or agents that can delay, or prevent AD and its progression, and we still do not have early detectable biomarkers for AD. Multiple cellular changes have been implicated in AD, including synaptic damage, mitochondrial damage, production and accumulation of Aβ and phosphorylated tau, inflammatory response, deficits in neurotransmitters, deregulation of the cell cycle, and hormonal imbalance. Research into AD has revealed that miRNAs are involved in each of these cellular changes and interfere with gene regulation and translation. Recent discoveries in molecular biology have also revealed that microRNAs play a major role in post-translational regulation of gene expression. The purpose of this article is to review research that has assessed neuroprotective and neurodegenerative characteristics of microRNAs in brain samples from AD transgenic mouse models and patients with AD.
Collapse
Affiliation(s)
- P Hemachandra Reddy
- Garrison Institute on Aging Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Cell Biology & Biochemistry Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Neuroscience & Pharmacology Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Neurology Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Speech, Language and Hearing Sciences Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, 6630 S. Quaker Suite E, MS 7495, Lubbock, TX 79413, United States.
| | - Sahil Tonk
- Garrison Institute on Aging Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States
| | - Subodh Kumar
- Garrison Institute on Aging Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States
| | - Murali Vijayan
- Garrison Institute on Aging Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States
| | - Ramesh Kandimalla
- Garrison Institute on Aging Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States
| | - Chandra Sekhar Kuruva
- Garrison Institute on Aging Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States
| | - Arubala P Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 Fourth Street, MS 9424, Lubbock, TX 79430, United States.
| |
Collapse
|
118
|
MicroRNAs underlying memory deficits in neurodegenerative disorders. Prog Neuropsychopharmacol Biol Psychiatry 2017; 73:79-86. [PMID: 27117821 DOI: 10.1016/j.pnpbp.2016.04.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/01/2016] [Accepted: 04/22/2016] [Indexed: 11/23/2022]
Abstract
Neurodegenerative disorders are defined by neuronal loss and often associated with dementia. Understanding the multifactorial nature of cognitive decline is of particular interest. Cell loss is certainly a possibility but also an early imbalance in the complex gene networks involved in learning and memory. The small (~22nt) non-coding microRNAs play a major role in gene expression regulation and have been linked to neuronal survival and cognition. Interestingly, changes in microRNA signatures are associated with neurodegenerative disorders. In this review, we explore the role of three microRNAs, namely miR-132, miR-124 and miR-34, which are dysregulated in major neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and Huntington's disease. Interestingly, these microRNAs have been associated with both memory impairment and neuronal survival, providing a potential common molecular mechanism contributing to dementia.
Collapse
|
119
|
Shah P, Cho SK, Thulstrup PW, Bjerrum MJ, Lee PH, Kang JH, Bhang YJ, Yang SW. MicroRNA Biomarkers in Neurodegenerative Diseases and Emerging Nano-Sensors Technology. J Mov Disord 2017; 10:18-28. [PMID: 28122423 PMCID: PMC5288660 DOI: 10.14802/jmd.16037] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 11/12/2016] [Accepted: 11/22/2016] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRNAs) are essential small RNA molecules (20–24 nt) that negatively regulate the expression of target genes at the post-transcriptional level. Due to their roles in a variety of biological processes, the aberrant expression profiles of miRNAs have been identified as biomarkers for many diseases, such as cancer, diabetes, cardiovascular disease and neurodegenerative diseases. In order to precisely, rapidly and economically monitor the expression of miRNAs, many cutting-edge nanotechnologies have been developed. One of the nanotechnologies, based on DNA encapsulated silver nanoclusters (DNA/AgNCs), has increasingly been adopted to create nanoscale bio-sensing systems due to its attractive optical properties, such as brightness, tuneable emission wavelengths and photostability. Using the DNA/AgNCs sensor methods, the presence of miRNAs can be detected simply by monitoring the fluorescence alteration of DNA/AgNCs sensors. We introduce these DNA/ AgNCs sensor methods and discuss their possible applications for detecting miRNA biomarkers in neurodegenerative diseases.
Collapse
Affiliation(s)
- Pratik Shah
- UNIK Center for Synthetic Biology, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Seok Keun Cho
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | | | | | - Phil Hyu Lee
- Department of Neurology, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ju-Hee Kang
- Department of Pharmacology, Hypoxia-related Disease Research Center, Inha University School of Medicine, Incheon, Korea
| | | | - Seong Wook Yang
- UNIK Center for Synthetic Biology, University of Copenhagen, Copenhagen, Denmark.,Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| |
Collapse
|
120
|
Yagishita S, Suzuki S, Yoshikawa K, Iida K, Hirata A, Suzuki M, Takashima A, Maruyama K, Hirasawa A, Awaji T. Treatment of intermittent hypoxia increases phosphorylated tau in the hippocampus via biological processes common to aging. Mol Brain 2017; 10:2. [PMID: 28057021 PMCID: PMC5217192 DOI: 10.1186/s13041-016-0282-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/15/2016] [Indexed: 12/17/2022] Open
Abstract
Sleep-disordered breathing produces cognitive impairments, and is possibly associated with Alzheimer disease (AD). Intermittent hypoxia treatment (IHT), an experimental model for sleep-disordered breathing, results in cognitive impairments in animals via unknown mechanisms. Here, we exposed mice to IHT protocols, and performed biochemical analyses and microarray analyses regarding their hippocampal samples. In particular, we performed gene ontology (GO)-based microarray analysis to elucidate effects of IHT on hippocampal functioning, which were compared with the effects of various previously-reported experimental conditions on that (ref. Gene Expression Omnibus, The National Center for Biotechnology Information). Our microarray analyses revealed that IHT and aging shared alterations in some common GO, which were also observed with kainic acid treatment, Dicer ablation, or moderate glutamate excess. Mapping the altered genes using the Kyoto Encyclopedia of Genes and Genomes PATHWAY database indicated that IHT and aging affected several pathways including “MAPK signaling pathway”, “PI3K-Akt signaling pathway”, and “glutamatergic synapse”. Consistent with the gene analyses, in vivo analyses revealed that IHT increased phosphorylated tau, reflecting an imbalance of kinases and/or phosphatases, and reduced proteins relevant to glutamatergic synapses. In addition, IHT increased phosphorylated p70 S6 kinase, indicating involvement of the mammalian target of rapamycin signaling pathway. Furthermore, IHT mice demonstrated hyperactivity in Y-maze tests, which was also observed in AD models. We obtained important data or something from the massive amount of microarray data, and confirmed the validity by in vivo analyses: the IHT-induced cognitive impairment may be partially explained by the fact that IHT increases phosphorylated tau via biological processes common to aging. Moreover, as aging is a major risk factor for AD, IHT is a novel model for investigating the pathological processes contributing to AD onset.
Collapse
Affiliation(s)
- Sosuke Yagishita
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan. .,Present address: Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.
| | - Seiya Suzuki
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan.,Faculty of Health and Medical Care, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
| | - Keisuke Yoshikawa
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan
| | - Keiko Iida
- Department of Genomic Drug Discovery Science, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Ayako Hirata
- Department of Genomic Drug Discovery Science, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Masahiko Suzuki
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan.,Faculty of Health and Medical Care, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1241, Japan
| | - Akihiko Takashima
- Department of Life Science, Faculty of Science, Gakushuin University, 1-5-1 Mejiro, Toshima-ku, Tokyo, 171-8588, Japan
| | - Kei Maruyama
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan
| | - Akira Hirasawa
- Department of Genomic Drug Discovery Science, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Takeo Awaji
- Department of Pharmacology, Faculty of Medicine, Saitama Medical University, 38 Moro-hongo, Moroyama-machi, Iruma-gun, Saitama, 350-0495, Japan
| |
Collapse
|
121
|
Reddy P, Williams J, Smith F, Bhatti J, Kumar S, Vijayan M, Kandimalla R, Kuruva C, Wang R, Manczak M, Yin X, Reddy A. MicroRNAs, Aging, Cellular Senescence, and Alzheimer's Disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 146:127-171. [PMID: 28253983 DOI: 10.1016/bs.pmbts.2016.12.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
122
|
|
123
|
Quinlan S, Kenny A, Medina M, Engel T, Jimenez-Mateos EM. MicroRNAs in Neurodegenerative Diseases. MIRNAS IN AGING AND CANCER 2017; 334:309-343. [DOI: 10.1016/bs.ircmb.2017.04.002] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
124
|
Pereira VG, Queiroz MT, D'Almeida V. Differential expression of microRNAs from miR-17 family in the cerebellum of mucopolysaccharidosis type I mice. Gene 2016; 595:207-211. [DOI: 10.1016/j.gene.2016.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/03/2016] [Accepted: 10/05/2016] [Indexed: 12/21/2022]
|
125
|
Wang X, Yin H, Rich AM, Sun Y, Tan Z, Luo X, Che N, Wei M, Yin J. MicroRNAs as biomarkers in molecular diagnosis of refractory epilepsy. Chin Neurosurg J 2016. [DOI: 10.1186/s41016-016-0049-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
126
|
Mufson EJ, Ikonomovic MD, Counts SE, Perez SE, Malek-Ahmadi M, Scheff SW, Ginsberg SD. Molecular and cellular pathophysiology of preclinical Alzheimer's disease. Behav Brain Res 2016; 311:54-69. [PMID: 27185734 PMCID: PMC4931948 DOI: 10.1016/j.bbr.2016.05.030] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/12/2016] [Accepted: 05/12/2016] [Indexed: 12/19/2022]
Abstract
Although the two pathological hallmarks of Alzheimer's disease (AD), senile plaques composed of amyloid-β (Aβ) peptides and neurofibrillary tangles (NFTs) consisting of hyperphosphorylated tau, have been studied extensively in postmortem AD and relevant animal and cellular models, the pathogenesis of AD remains unknown, particularly in the early stages of the disease where therapies presumably would be most effective. We and others have demonstrated that Aβ plaques and NFTs are present in varying degrees before the onset and throughout the progression of dementia. In this regard, aged people with no cognitive impairment (NCI), mild cognitive impairment (MCI, a presumed prodromal AD transitional state, and AD all present at autopsy with varying levels of pathological hallmarks. Cognitive decline, a requisite for the clinical diagnosis of dementia associated with AD, generally correlates better with NFTs than Aβ plaques. However, correlations are even higher between cognitive decline and synaptic loss. In this review, we illustrate relevant clinical pathological research in preclinical AD and throughout the progression of dementia in several areas including Aβ and tau pathobiology, single population expression profiling of vulnerable hippocampal and basal forebrain neurons, neuroplasticity, neuroimaging, cerebrospinal fluid (CSF) biomarker studies and their correlation with antemortem cognitive endpoints. In each of these areas, we provide evidence for the importance of studying the pathological hallmarks of AD not in isolation, but rather in conjunction with other molecular, cellular, and imaging markers to provide a more systematic and comprehensive assessment of the multiple changes that occur during the transition from NCI to MCI to frank AD.
Collapse
Affiliation(s)
- Elliott J Mufson
- Departments of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ, United States.
| | - Milos D Ikonomovic
- Departments of Neurology and Psychiatry, University of Pittsburgh, and Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, United States
| | - Scott E Counts
- Department of Translational Science and Molecular Medicine, Department of Family Medicine, Hauenstien Neuroscience Institute, Mercy Health Saint Mary's Hospital, Grand Rapids, MI, United States
| | - Sylvia E Perez
- Departments of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ, United States
| | | | - Stephen W Scheff
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Department of Psychiatry, Department of Neuroscience & Physiology, New York University Langone Medical Center, Orangeburg, NY, United States
| |
Collapse
|
127
|
Jiao Y, Kong L, Yao Y, Li S, Tao Z, Yan Y, Yang J. Osthole decreases beta amyloid levels through up-regulation of miR-107 in Alzheimer’s disease. Neuropharmacology 2016; 108:332-44. [DOI: 10.1016/j.neuropharm.2016.04.046] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 04/07/2016] [Accepted: 04/29/2016] [Indexed: 11/27/2022]
|
128
|
Jauhari A, Singh T, Pandey A, Singh P, Singh N, Srivastava AK, Pant AB, Parmar D, Yadav S. Differentiation Induces Dramatic Changes in miRNA Profile, Where Loss of Dicer Diverts Differentiating SH-SY5Y Cells Toward Senescence. Mol Neurobiol 2016; 54:4986-4995. [PMID: 27525675 DOI: 10.1007/s12035-016-0042-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/05/2016] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) are generated by endonuclease activity of Dicer, which also helps in loading of miRNAs to their target sequences. SH-SY5Y, a human neuroblastoma and a cellular model of neurodevelopment, consistently expresses genes related to neurodegenerative disorders at different biological levels (DNA, RNA, and proteins). Using SH-SY5Y cells, we have studied the role of Dicer and miRNAs in neuronal differentiation and explored involvement of P53, a master regulator of gene expression in differentiation-induced induction of miRNAs. Knocking down Dicer gene induced senescence in differentiating SH-SY5Y cells, which indicate the essential role of Dicer in brain development. Differentiation of SH-SY5Y cells by retinoic acid (RA) or RA + brain-derived neurotrophic factor (BDNF) induced dramatic changes in global miRNA expression. Fully differentiated SH-SY5Y cells (5-day RA followed by 3-day BDNF) significantly (p < 0.05 and atleast >3-fold change) upregulated and downregulated the expression of 77 and 17 miRNAs, respectively. Maximum increase was observed in the expression of miR-193-5p, miR-199a-5p, miR-192, miR-145, miR-28-5p, miR-29b, and miR-222 after RA exposure and miR-193-5p, miR-146a, miR-21, miR-199a-5p, miR-153, miR-29b, and miR-222 after RA + BDNF exposure in SH-SY5Y cells. Exploring the role of P53 in differentiating SH-SY5Y cells, we have observed that induction of miR-222, miR-192, and miR-145 is P53 dependent and expression of miR-193a-5p, miR-199a-5p, miR-146a, miR-21, miR-153, and miR-29b is P53 independent. In conclusion, decreased Dicer level enforces differentiating cells to senescence, and differentiating SH-SY5Y cells needs increased expression of P53 to cope up with changes in protein levels of mature neurons.
Collapse
Affiliation(s)
- Abhishek Jauhari
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (IITR), Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow, UP, 226001, India.,Academy of Scientific and Innovative Research (AcSIR), IITR Campus, Lucknow, India
| | - Tanisha Singh
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (IITR), Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow, UP, 226001, India.,Department of Biochemistry, School of Dental Sciences, Babu Banarasi Das University, Lucknow, India
| | - Ankita Pandey
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (IITR), Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow, UP, 226001, India
| | - Parul Singh
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (IITR), Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow, UP, 226001, India.,Department of Biochemistry, School of Dental Sciences, Babu Banarasi Das University, Lucknow, India
| | - Nishant Singh
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (IITR), Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow, UP, 226001, India.,Department of Biochemistry, School of Dental Sciences, Babu Banarasi Das University, Lucknow, India
| | - Ankur Kumar Srivastava
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (IITR), Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow, UP, 226001, India.,Academy of Scientific and Innovative Research (AcSIR), IITR Campus, Lucknow, India
| | - Aditya Bhushan Pant
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (IITR), Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow, UP, 226001, India
| | - Devendra Parmar
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (IITR), Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow, UP, 226001, India
| | - Sanjay Yadav
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (IITR), Vishvigyan Bhawan, 31 Mahatma Gandhi Marg, Lucknow, UP, 226001, India.
| |
Collapse
|
129
|
Karnati HK, Panigrahi MK, Gutti RK, Greig NH, Tamargo IA. miRNAs: Key Players in Neurodegenerative Disorders and Epilepsy. J Alzheimers Dis 2016; 48:563-80. [PMID: 26402105 DOI: 10.3233/jad-150395] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) are endogenous, ∼22 nucleotide, non-coding RNA molecules that function as post-transcriptional regulators of gene expression. miRNA dysregulation has been observed in cancer and in neurodegenerative disorders such as Alzheimer's, Parkinson's, and Huntington's diseases, amyotrophic lateral sclerosis, and the neurological disorder, epilepsy. Neuronal degradation and death are important hallmarks of neurodegenerative disorders. Additionally, abnormalities in metabolism, synapsis and axonal transport have been associated with Alzheimer's disease, Parkinson's disease, and frontotemporal dementia. A number of recently published studies have demonstrated the importance of miRNAs in the nervous system and have contributed to the growing body of evidence on miRNA dysregulation in neurological disorders. Knowledge of the expressions and activities of such miRNAs may aid in the development of novel therapeutics. In this review, we discuss the significance of miRNA dysregulation in the development of neurodegenerative disorders and the use of miRNAs as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Hanuma Kumar Karnati
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, Telangana, India
| | - Manas Kumar Panigrahi
- Department of Neurosurgery, Krishna Institute of Medical Sciences (KIMS), Hyderabad, Telangana, India
| | - Ravi Kumar Gutti
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, Telangana, India
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ian A Tamargo
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
130
|
Di Meco A, Praticò D. MicroRNAs as Therapeutic Targets for Alzheimer’s Disease. J Alzheimers Dis 2016; 53:367-72. [DOI: 10.3233/jad-160203] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
131
|
Zhang B, Wang LL, Ren RJ, Dammer EB, Zhang YF, Huang Y, Chen SD, Wang G. MicroRNA-146a represses LRP2 translation and leads to cell apoptosis in Alzheimer's disease. FEBS Lett 2016; 590:2190-200. [PMID: 27241555 DOI: 10.1002/1873-3468.12229] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/10/2016] [Accepted: 05/24/2016] [Indexed: 12/17/2022]
Abstract
MicroRNA regulation of transcript expression has been reported in patients with Alzheimer's disease (AD). Here, we investigate the role of microRNA-146a (miRNA-146a), a brain-enriched miRNA, which is upregulated in AD patients. Through analysis of predicted targets of miRNA-146a, low-density lipoprotein receptor-related protein-2 (Lrp2), a member of the LDLR family that is known to play a protective role in AD, was identified. Overexpression of miRNA-146a in SH-SY5Y cells significantly decreased Lrp2 expression, resulting in a reduction of Akt activation and induction of proapoptotic caspase-3, thereby increasing cell apoptosis. Thus, specific miRNA-146a regulation may contribute to AD by downregulating the Lrp2/Akt pathway.
Collapse
Affiliation(s)
- Bei Zhang
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China.,Laboratory of Neurodegenerative Diseases, Institute of Health Science, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, China
| | - Li-Ling Wang
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Ru-Jing Ren
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China
| | - Eric B Dammer
- Department of Biochemistry and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Yong-Fang Zhang
- Research Laboratory of Cell Regulation, Shanghai Jiao Tong University School of Medicine, China
| | - Yue Huang
- School of Medical Sciences, The University of New South Wales, Randwick, Australia
| | - Sheng-Di Chen
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China.,Laboratory of Neurodegenerative Diseases, Institute of Health Science, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, China
| | - Gang Wang
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
132
|
Moncini S, Lunghi M, Valmadre A, Grasso M, Del Vescovo V, Riva P, Denti MA, Venturin M. The miR-15/107 Family of microRNA Genes Regulates CDK5R1/p35 with Implications for Alzheimer's Disease Pathogenesis. Mol Neurobiol 2016; 54:4329-4342. [PMID: 27343180 DOI: 10.1007/s12035-016-0002-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 06/14/2016] [Indexed: 02/06/2023]
Abstract
Cyclin-dependent kinase 5 regulatory subunit 1 (CDK5R1) encodes p35, the main activatory subunit of cyclin-dependent kinase 5 (CDK5). The p35/CDK5 active complex plays a fundamental role in brain development and functioning, but its deregulated activity has also been implicated in various neurodegenerative disorders, including Alzheimer's disease (AD). CDK5R1 displays a large and highly evolutionarily conserved 3'-untranslated region (3'-UTR), a fact that has suggested a role for this region in the post-transcriptional control of CDK5R1 expression. Our group has recently demonstrated that two miRNAs, miR-103 and miR-107, regulate CDK5R1 expression and affect the levels of p35. MiR-103 and miR-107 belong to the miR-15/107 family, a group of evolutionarily conserved miRNAs highly expressed in human cerebral cortex. In this work, we tested the hypothesis that other members of this group of miRNAs, in addition to miR-103 and miR-107, were able to modulate CDK5R1 expression. We provide evidence that several miRNAs belonging to the miR-15/107 family regulate p35 levels. BACE1 expression levels were also found to be modulated by different members of this family. Furthermore, overexpression of these miRNAs led to reduced APP phosphorylation levels at the CDK5-specific Thr668 residue. We also show that miR-15/107 miRNAs display reduced expression levels in hippocampus and temporal cortex, but not in cerebellum, of AD brains. Moreover, increased CDK5R1 mRNA levels were observed in AD hippocampus tissues. Our results suggest that the downregulation of the miR-15/107 family might have a role in the pathogenesis of AD by increasing the levels of CDK5R1/p35 and consequently enhancing CDK5 activity.
Collapse
Affiliation(s)
- Silvia Moncini
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Via Viotti 3/5, 20133, Milan, Italy
| | - Marta Lunghi
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Via Viotti 3/5, 20133, Milan, Italy
| | - Alice Valmadre
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Via Viotti 3/5, 20133, Milan, Italy
| | - Margherita Grasso
- Centre for Integrative Biology, Università degli Studi di Trento, Via Sommarive 9, 38123, Povo, (TN), Italy
| | - Valerio Del Vescovo
- Centre for Integrative Biology, Università degli Studi di Trento, Via Sommarive 9, 38123, Povo, (TN), Italy
| | - Paola Riva
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Via Viotti 3/5, 20133, Milan, Italy
| | - Michela Alessandra Denti
- Centre for Integrative Biology, Università degli Studi di Trento, Via Sommarive 9, 38123, Povo, (TN), Italy.,Istituto di Neuroscienze, CNR, Viale Giuseppe Colombo 3, 35121, Padova, Italy
| | - Marco Venturin
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Via Viotti 3/5, 20133, Milan, Italy.
| |
Collapse
|
133
|
Hollins SL, Cairns MJ. MicroRNA: Small RNA mediators of the brains genomic response to environmental stress. Prog Neurobiol 2016; 143:61-81. [PMID: 27317386 DOI: 10.1016/j.pneurobio.2016.06.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/24/2016] [Accepted: 06/11/2016] [Indexed: 01/09/2023]
Abstract
The developmental processes that establish the synaptic architecture of the brain while retaining capacity for activity-dependent remodeling, are complex and involve a combination of genetic and epigenetic influences. Dysregulation of these processes can lead to problems with neural circuitry which manifest in humans as a range of neurodevelopmental syndromes, such as schizophrenia, bipolar disorder and fragile X mental retardation. Recent studies suggest that prenatal, postnatal and intergenerational environmental factors play an important role in the aetiology of stress-related psychopathology. A number of these disorders have been shown to display epigenetic changes in the postmortem brain that reflect early life experience. These changes affect the regulation of gene expression though chromatin remodeling (transcriptional) and post-transcriptional influences, especially small noncoding microRNA (miRNA). These dynamic and influential molecules appear to play an important function in both brain development and its adaption to stress. In this review, we examine the role of miRNA in mediating the brain's response to both prenatal and postnatal environmental perturbations and explore how stress- induced alterations in miRNA expression can regulate the stress response via modulation of the immune system. Given the close relationship between environmental stress, miRNA, and brain development/function, we assert that miRNA hold a significant position at the molecular crossroads between neural development and adaptations to environmental stress. A greater understanding of the dynamics that mediate an individual's predisposition to stress-induced neuropathology has major human health benefits and is an important area of research.
Collapse
Affiliation(s)
- Sharon L Hollins
- School of Biomedical Sciences and Pharmacy and the Hunter Medical Research Institute, the University of Newcastle, Callaghan, NSW 2308, Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy and the Hunter Medical Research Institute, the University of Newcastle, Callaghan, NSW 2308, Australia; Schizophrenia Research Institute, Sydney, NSW, Australia.
| |
Collapse
|
134
|
Wang T, Shi F, Jin Y, Jiang W, Shen D, Xiao S. Abnormal Changes of Brain Cortical Anatomy and the Association with Plasma MicroRNA107 Level in Amnestic Mild Cognitive Impairment. Front Aging Neurosci 2016; 8:112. [PMID: 27242521 PMCID: PMC4870937 DOI: 10.3389/fnagi.2016.00112] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 04/29/2016] [Indexed: 11/17/2022] Open
Abstract
UNLABELLED MicroRNA107 (Mir107) has been thought to relate to the brain structure phenotype of Alzheimer's disease. In this study, we evaluated the cortical anatomy in amnestic mild cognitive impairment (aMCI) and the relation between cortical anatomy and plasma levels of Mir107 and beta-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1). Twenty aMCI (20 aMCI) and 24 cognitively normal control (NC) subjects were recruited, and T1-weighted MR images were acquired. Cortical anatomical measurements, including cortical thickness (CT), surface area (SA), and local gyrification index (LGI), were assessed. Quantitative RT-PCR was used to examine plasma expression of Mir107, BACE1 mRNA. Thinner cortex was found in aMCI in areas associated with episodic memory and language, but with thicker cortex in other areas. SA decreased in aMCI in the areas associated with working memory and emotion. LGI showed a significant reduction in aMCI in the areas involved in language function. Changes in Mir107 and BACE1 messenger RNA plasma expression were correlated with changes in CT and SA. We found alterations in key left brain regions associated with memory, language, and emotion in aMCI that were significantly correlated with plasma expression of Mir107 and BACE1 mRNA. This combination study of brain anatomical alterations and gene information may shed lights on our understanding of the pathology of AD. CLINICAL TRIAL REGISTRATION http://www.ClinicalTrials.gov, identifier NCT01819545.
Collapse
Affiliation(s)
- Tao Wang
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of MedicineShanghai, China
- Alzheimer’s Disease and Related Disorders Center, Shanghai Jiao Tong UniversityShanghai, China
- IDEA Lab, Department of Radiology and BRIC, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Feng Shi
- IDEA Lab, Department of Radiology and BRIC, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Yan Jin
- IDEA Lab, Department of Radiology and BRIC, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Weixiong Jiang
- Alzheimer’s Disease and Related Disorders Center, Shanghai Jiao Tong UniversityShanghai, China
| | - Dinggang Shen
- IDEA Lab, Department of Radiology and BRIC, University of North Carolina at Chapel HillChapel Hill, NC, USA
- Department of Brain and Cognitive Engineering, Korea UniversitySeoul, South Korea
| | - Shifu Xiao
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of MedicineShanghai, China
- Alzheimer’s Disease and Related Disorders Center, Shanghai Jiao Tong UniversityShanghai, China
| |
Collapse
|
135
|
Fiorenza A, Barco A. Role of Dicer and the miRNA system in neuronal plasticity and brain function. Neurobiol Learn Mem 2016; 135:3-12. [PMID: 27163737 DOI: 10.1016/j.nlm.2016.05.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/05/2016] [Accepted: 05/05/2016] [Indexed: 01/26/2023]
Abstract
MicroRNAs (miRNAs) are small regulatory non-coding RNAs that contribute to fine-tuning regulation of gene expression by mRNA destabilization and/or translational repression. Their abundance in the nervous system, their temporally and spatially regulated expression and their ability to respond in an activity-dependent manner make miRNAs ideal candidates for the regulation of complex processes in the brain, including neuronal plasticity, memory formation and neural development. The conditional ablation of the RNase III Dicer, which is essential for the maturation of most miRNAs, is a useful model to investigate the effect of the loss of the miRNA system, as a whole, in different tissues and cellular types. In this review, we first provide an overview of Dicer function and structure, and discuss outstanding questions concerning the role of miRNAs in the regulation of gene expression and neuronal function, to later focus on the insight derived from studies in which the genetic ablation of Dicer was used to determine the role of the miRNA system in the nervous system. In particular, we highlight the collective role of miRNAs fine-tuning plasticity-related gene expression and providing robustness to neuronal gene expression networks.
Collapse
Affiliation(s)
- Anna Fiorenza
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant, 03550 Alicante, Spain
| | - Angel Barco
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant, 03550 Alicante, Spain.
| |
Collapse
|
136
|
Magner WJ, Weinstock-Guttman B, Rho M, Hojnacki D, Ghazi R, Ramanathan M, Tomasi TB. Dicer and microRNA expression in multiple sclerosis and response to interferon therapy. J Neuroimmunol 2016; 292:68-78. [PMID: 26943961 PMCID: PMC4779496 DOI: 10.1016/j.jneuroim.2016.01.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/06/2016] [Accepted: 01/14/2016] [Indexed: 12/11/2022]
Abstract
Dysregulation of microRNA expression has been shown in multiple sclerosis (MS); however, the mechanisms underlying these changes, their response to therapy and the impact of microRNA changes in MS are not completely understood. Dicer mediates the cleavage of precursor microRNAs to mature microRNAs and is dysregulated in multiple pathologies. Having shown that interferons regulate Dicer in vitro, we hypothesized that MS patient IFNβ1a treatment could potentially alter Dicer expression. Dicer mRNA and protein levels, as well as microRNA expression, were determined in MS patient and healthy control PBL. Acute responses to IFNβ1a were assessed in 50 patients. We found that Dicer protein but not mRNA levels decreases in MS patients while both are selectively induced in patients responding well to IFNβ1a. Potential microRNA biomarkers for relapsing remitting multiple sclerosis (RRMS), secondary progressive multiple sclerosis (SPMS) and IFNβ1a response are described. Contrasts in Dicer and microRNA expression levels between patient populations may offer insight into mechanisms underlying disease courses and responses to IFNβ1a therapy. This work identifies Dicer regulation as both a potential mediator of MS pathology and a therapeutic target.
Collapse
Affiliation(s)
- William J Magner
- Laboratory of Molecular Medicine, Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA; Department of Microbiology and Immunology, State University of New York, School of Medicine and Biomedical Sciences, Buffalo, NY, USA.
| | - Bianca Weinstock-Guttman
- Jacobs Neurological Institute, Buffalo, NY, USA; Department of Neurology, State University of New York, School of Medicine and Biomedical Sciences, Buffalo, NY, USA.
| | - Mina Rho
- Division of Computer Science and Engineering, Hanyang University, Seoul, Republic of Korea.
| | - David Hojnacki
- Jacobs Neurological Institute, Buffalo, NY, USA; Department of Neurology, State University of New York, School of Medicine and Biomedical Sciences, Buffalo, NY, USA.
| | - Rabia Ghazi
- Jacobs Neurological Institute, Buffalo, NY, USA; Department of Neurology, State University of New York, School of Medicine and Biomedical Sciences, Buffalo, NY, USA.
| | - Murali Ramanathan
- Jacobs Neurological Institute, Buffalo, NY, USA; Department of Neurology, State University of New York, School of Medicine and Biomedical Sciences, Buffalo, NY, USA; Department of Pharmaceutical Sciences, State University of New York, School of Medicine and Biomedical Sciences, Buffalo, NY, USA.
| | - Thomas B Tomasi
- Laboratory of Molecular Medicine, Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA; Department of Microbiology and Immunology, State University of New York, School of Medicine and Biomedical Sciences, Buffalo, NY, USA; Department of Medicine, State University of New York, School of Medicine and Biomedical Sciences, Buffalo, NY, USA.
| |
Collapse
|
137
|
Epithelium-Intrinsic MicroRNAs Contribute to Mucosal Immune Homeostasis by Promoting M-Cell Maturation. PLoS One 2016; 11:e0150379. [PMID: 26930511 PMCID: PMC4773159 DOI: 10.1371/journal.pone.0150379] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/13/2016] [Indexed: 12/24/2022] Open
Abstract
M cells in the follicle-associated epithelium (FAE) of Peyer’s patches (PPs) serve as a main portal for external antigens and function as a sentinel in mucosal immune responses. The scarcity of these cells has hampered identification of M cell-specific molecules. Recent efforts have begun to provide insight into antigen transcytosis and differentiation of M cells; however, the molecular mechanisms underlying these processes are not fully elucidated. Small non-coding RNAs including microRNA (miRNA) have been reported to regulate gene expression and control various biological processes such as cellular differentiation and function. To evaluate the expression of miRNAs in FAE, including M cells, we previously performed microarray analysis comparing intestinal villous epithelium (VE) and PP FAE. Here we confirmed FAE specific miRNA expression levels by quantitative PCR. To gain insight into miRNA function, we generated mice with intestinal epithelial cell-specific deletion of Dicer1 (DicerΔIEC) and analyzed intestinal phenotypes, including M-cell differentiation, morphology and function. DicerΔIEC mice had a marked decrease in M cells compared to control floxed Dicer mice, suggesting an essential role of miRNAs in maturation of these cells. Furthermore, transmission electron microscopic analysis revealed that depletion of miRNA caused the loss of endosomal structures in M cells. In addition, antigen uptake by M cells was impaired in DicerΔIEC mice. These results suggest that miRNAs play a significant role in M cell differentiation and help secure mucosal immune homeostasis.
Collapse
|
138
|
Hu YB, Li CB, Song N, Zou Y, Chen SD, Ren RJ, Wang G. Diagnostic Value of microRNA for Alzheimer's Disease: A Systematic Review and Meta-Analysis. Front Aging Neurosci 2016; 8:13. [PMID: 26903857 PMCID: PMC4746262 DOI: 10.3389/fnagi.2016.00013] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/15/2016] [Indexed: 01/12/2023] Open
Abstract
Sound evidence indicates that microRNAs (miRNAs) are aberrantly expressed in Alzheimer's disease (AD) patients. We performed a systematic review and meta-analysis to investigate the role of miRNA in AD pathogenesis and their clinical diagnostic value; a systematic review of literature and meta-analysis of clinical trials were performed. In the systematic review, 236 papers were included, and we reviewed the dysregulated miRNA expression in different parts of AD patients in order to identify the relationship between aberrantly expressed miRNAs and AD pathology. In the subsequent meta-analysis, seven studies were statistically analyzed with the following results: pooled sensitivity 0.86 (95%CI 0.79-0.90), pooled specificity 0.87 (95%CI 0.72-0.95), diagnostic odds ratio (28.29), and the area under the curve (0.87). In conclusion, our review indicated that aberrant expression of various miRNAs plays an important role in the pathological process of AD, and statistical analysis of quantitative studies reveal the potential value of specific miRNAs in the diagnosis of AD.
Collapse
Affiliation(s)
- Yong-Bo Hu
- Department of Neurology, Neuroscience Institute, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Chun-Bo Li
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Ning Song
- St George Hospital , Sydney, NSW , Australia
| | - Yang Zou
- Department of Neurology, Neuroscience Institute, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Sheng-Di Chen
- Department of Neurology, Neuroscience Institute, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Ru-Jing Ren
- Department of Neurology, Neuroscience Institute, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Gang Wang
- Department of Neurology, Neuroscience Institute, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| |
Collapse
|
139
|
Malmevik J, Petri R, Knauff P, Brattås PL, Åkerblom M, Jakobsson J. Distinct cognitive effects and underlying transcriptome changes upon inhibition of individual miRNAs in hippocampal neurons. Sci Rep 2016; 6:19879. [PMID: 26813637 PMCID: PMC4728481 DOI: 10.1038/srep19879] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/21/2015] [Indexed: 11/09/2022] Open
Abstract
MicroRNAs (miRNA) are small, non-coding RNAs mediating post-transcriptional regulation of gene expression. miRNAs have recently been implicated in hippocampus-dependent functions such as learning and memory, although the roles of individual miRNAs in these processes remain largely unknown. Here, we achieved stable inhibition using AAV-delivered miRNA sponges of individual, highly expressed and brain-enriched miRNAs; miR-124, miR-9 and miR-34, in hippocampal neurons. Molecular and cognitive studies revealed a role for miR-124 in learning and memory. Inhibition of miR-124 resulted in an enhanced spatial learning and working memory capacity, potentially through altered levels of genes linked to synaptic plasticity and neuronal transmission. In contrast, inhibition of miR-9 or miR-34 led to a decreased capacity of spatial learning and of reference memory, respectively. On a molecular level, miR-9 inhibition resulted in altered expression of genes related to cell adhesion, endocytosis and cell death, while miR-34 inhibition caused transcriptome changes linked to neuroactive ligand-receptor transduction and cell communication. In summary, this study establishes distinct roles for individual miRNAs in hippocampal function.
Collapse
Affiliation(s)
- Josephine Malmevik
- Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, Sölvegatan 17, 221 84 Lund, Sweden
| | - Rebecca Petri
- Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, Sölvegatan 17, 221 84 Lund, Sweden
| | - Pina Knauff
- Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, Sölvegatan 17, 221 84 Lund, Sweden
| | - Per Ludvik Brattås
- Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, Sölvegatan 17, 221 84 Lund, Sweden
| | - Malin Åkerblom
- Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, Sölvegatan 17, 221 84 Lund, Sweden
| | - Johan Jakobsson
- Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, BMC A11, Lund University, Sölvegatan 17, 221 84 Lund, Sweden
| |
Collapse
|
140
|
Roth W, Hecker D, Fava E. Systems Biology Approaches to the Study of Biological Networks Underlying Alzheimer's Disease: Role of miRNAs. Methods Mol Biol 2016; 1303:349-377. [PMID: 26235078 DOI: 10.1007/978-1-4939-2627-5_21] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
MicroRNAs (miRNAs) are emerging as significant regulators of mRNA complexity in the human central nervous system (CNS) thereby controlling distinct gene expression profiles in a spatio-temporal manner during development, neuronal plasticity, aging and (age-related) neurodegeneration, including Alzheimer's disease (AD). Increasing effort is expended towards dissecting and deciphering the molecular and genetic mechanisms of neurobiological and pathological functions of these brain-enriched miRNAs. Along these lines, recent data pinpoint distinct miRNAs and miRNA networks being linked to APP splicing, processing and Aβ pathology (Lukiw et al., Front Genet 3:327, 2013), and furthermore, to the regulation of tau and its cellular subnetworks (Lau et al., EMBO Mol Med 5:1613, 2013), altogether underlying the onset and propagation of Alzheimer's disease. MicroRNA profiling studies in Alzheimer's disease suffer from poor consensus which is an acknowledged concern in the field, and constitutes one of the current technical challenges. Hence, a strong demand for experimental and computational systems biology approaches arises, to incorporate and integrate distinct levels of information and scientific knowledge into a complex system of miRNA networks in the context of the transcriptome, proteome and metabolome in a given cellular environment. Here, we will discuss the state-of-the-art technologies and computational approaches on hand that may lead to a deeper understanding of the complex biological networks underlying the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Wera Roth
- German Center for Neurodegenerative Diseases (DZNE), Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| | | | | |
Collapse
|
141
|
Liu Y, Yang X, Zhao L, Zhang J, Li T, Ma X. Increased miR-132 level is associated with visual memory dysfunction in patients with depression. Neuropsychiatr Dis Treat 2016; 12:2905-2911. [PMID: 27877044 PMCID: PMC5108558 DOI: 10.2147/ndt.s116287] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Impaired visual memory seems to be a core feature of depression, while increased microRNA-132 (miR-132) levels have been widely reported in depression patients. The authors aimed to explore the relationship between miR-132 changes and visual memory deficits in unmedicated patients with major depressive disorder (MDD). PATIENTS AND METHODS A total of 62 medication-free MDD patients and 73 matched healthy controls (HCs) were tested for miR-132 expression level in peripheral blood using quantitative real-time polymerase chain reaction. We used a computerized neurocognitive task from the Cambridge Neuropsychological Test Automated Battery (CANTAB) - pattern recognition memory (PRM) task - as a measurement of visual memory. The relationship between visual memory, miR-132 expression level, and clinical symptoms was explored in patients with MDD. RESULTS Upregulated miR-132 expression levels were seen in MDD patients but not in HCs. Two-sample t-tests showed that MDD patients had decreased visual memory, mainly memory delayed compared to that of HCs. Correlation analyses revealed that in MDD patients, increased miR-132 expression levels were significantly correlated with visual memory as measured by the CANTABPRM. Hamilton Rating Scale for Anxiety scores were negatively correlated with PRM - number correct (immediate) and PRM - percent correct (immediate). LIMITATIONS The main limitations were missing data and lack of follow-up studies. CONCLUSION Our study suggests that increased miR-132 expression levels were associated with visual memory deficits, which may underlie the pathophysiology of MDD. In individuals with depression, immediate visual memory defects were positively correlated with anxiety symptoms.
Collapse
Affiliation(s)
- Ye Liu
- Psychiatric Laboratory, Department of Psychiatry; National Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiao Yang
- Psychiatric Laboratory, Department of Psychiatry; National Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Liansheng Zhao
- Psychiatric Laboratory, Department of Psychiatry; National Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jian Zhang
- Psychiatric Laboratory, Department of Psychiatry; National Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tao Li
- Psychiatric Laboratory, Department of Psychiatry; National Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaohong Ma
- Psychiatric Laboratory, Department of Psychiatry; National Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
142
|
Swahari V, Nakamura A, Baran-Gale J, Garcia I, Crowther AJ, Sons R, Gershon TR, Hammond S, Sethupathy P, Deshmukh M. Essential Function of Dicer in Resolving DNA Damage in the Rapidly Dividing Cells of the Developing and Malignant Cerebellum. Cell Rep 2015; 14:216-24. [PMID: 26748703 DOI: 10.1016/j.celrep.2015.12.037] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 10/23/2015] [Accepted: 12/06/2015] [Indexed: 01/09/2023] Open
Abstract
Maintenance of genomic integrity is critical during neurodevelopment, particularly in rapidly dividing cerebellar granule neuronal precursors that experience constitutive replication-associated DNA damage. As Dicer was recently recognized to have an unexpected function in the DNA damage response, we examined whether Dicer was important for preserving genomic integrity in the developing brain. We report that deletion of Dicer in the developing mouse cerebellum resulted in the accumulation of DNA damage leading to cerebellar progenitor degeneration, which was rescued with p53 deficiency; deletion of DGCR8 also resulted in similar DNA damage and cerebellar degeneration. Dicer deficiency also resulted in DNA damage and death in other rapidly dividing cells including embryonic stem cells and the malignant cerebellar progenitors in a mouse model of medulloblastoma. Together, these results identify an essential function of Dicer in resolving the spontaneous DNA damage that occurs during the rapid proliferation of developmental progenitors and malignant cells.
Collapse
Affiliation(s)
- Vijay Swahari
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Ayumi Nakamura
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA; Neurobiology Curriculum, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jeanette Baran-Gale
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Idoia Garcia
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Neurology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Andrew J Crowther
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Neurology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Robert Sons
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Timothy R Gershon
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA; Neurobiology Curriculum, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Neurology, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Scott Hammond
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Praveen Sethupathy
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Mohanish Deshmukh
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA; Neurobiology Curriculum, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
143
|
Wood SH, van Dam S, Craig T, Tacutu R, O'Toole A, Merry BJ, de Magalhães JP. Transcriptome analysis in calorie-restricted rats implicates epigenetic and post-translational mechanisms in neuroprotection and aging. Genome Biol 2015; 16:285. [PMID: 26694192 PMCID: PMC4699360 DOI: 10.1186/s13059-015-0847-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/27/2015] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Caloric restriction (CR) can increase longevity in rodents and improve memory function in humans. α-Lipoic acid (LA) has been shown to improve memory function in rats, but not longevity. While studies have looked at survival in rodents after switching from one diet to another, the underlying mechanisms of the beneficial effects of CR and LA supplementation are unknown. Here, we use RNA-seq in cerebral cortex from rats subjected to CR and LA-supplemented rats to understand how changes in diet can affect aging, neurodegeneration and longevity. RESULTS Gene expression changes during aging in ad libitum-fed rats are largely prevented by CR, and neuroprotective genes are overexpressed in response to both CR and LA diets with a strong overlap of differentially expressed genes between the two diets. Moreover, a number of genes are differentially expressed specifically in rat cohorts exhibiting diet-induced life extension. Finally, we observe that LA supplementation inhibits histone deacetylase (HDAC) protein activity in vitro in rat astrocytes. We find a single microRNA, miR-98-3p, that is overexpressed during CR feeding and LA dietary supplementation; this microRNA alters HDAC and histone acetyltransferase (HAT) activity, which suggests a role for HAT/HDAC homeostasis in neuroprotection. CONCLUSIONS This study presents extensive data on the effects of diet and aging on the cerebral cortex transcriptome, and also emphasises the importance of epigenetics and post-translational modifications in longevity and neuroprotection.
Collapse
Affiliation(s)
- Shona H Wood
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Sipko van Dam
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Thomas Craig
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Robi Tacutu
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Amy O'Toole
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Brian J Merry
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
144
|
Cacabelos R, Torrellas C. Epigenetics of Aging and Alzheimer's Disease: Implications for Pharmacogenomics and Drug Response. Int J Mol Sci 2015; 16:30483-543. [PMID: 26703582 PMCID: PMC4691177 DOI: 10.3390/ijms161226236] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/16/2015] [Accepted: 12/08/2015] [Indexed: 02/07/2023] Open
Abstract
Epigenetic variability (DNA methylation/demethylation, histone modifications, microRNA regulation) is common in physiological and pathological conditions. Epigenetic alterations are present in different tissues along the aging process and in neurodegenerative disorders, such as Alzheimer’s disease (AD). Epigenetics affect life span and longevity. AD-related genes exhibit epigenetic changes, indicating that epigenetics might exert a pathogenic role in dementia. Epigenetic modifications are reversible and can potentially be targeted by pharmacological intervention. Epigenetic drugs may be useful for the treatment of major problems of health (e.g., cancer, cardiovascular disorders, brain disorders). The efficacy and safety of these and other medications depend upon the efficiency of the pharmacogenetic process in which different clusters of genes (pathogenic, mechanistic, metabolic, transporter, pleiotropic) are involved. Most of these genes are also under the influence of the epigenetic machinery. The information available on the pharmacoepigenomics of most drugs is very limited; however, growing evidence indicates that epigenetic changes are determinant in the pathogenesis of many medical conditions and in drug response and drug resistance. Consequently, pharmacoepigenetic studies should be incorporated in drug development and personalized treatments.
Collapse
Affiliation(s)
- Ramón Cacabelos
- EuroEspes Biomedical Research Center, Institute of Medical Science and Genomic Medicine, 15165-Bergondo, Corunna, Spain.
- Chair of Genomic Medicine, Camilo José Cela University, 28692-Madrid, Spain.
| | - Clara Torrellas
- EuroEspes Biomedical Research Center, Institute of Medical Science and Genomic Medicine, 15165-Bergondo, Corunna, Spain.
- Chair of Genomic Medicine, Camilo José Cela University, 28692-Madrid, Spain.
| |
Collapse
|
145
|
Smith PY, Hernandez-Rapp J, Jolivette F, Lecours C, Bisht K, Goupil C, Dorval V, Parsi S, Morin F, Planel E, Bennett DA, Fernandez-Gomez FJ, Sergeant N, Buée L, Tremblay MÈ, Calon F, Hébert SS. miR-132/212 deficiency impairs tau metabolism and promotes pathological aggregation in vivo. Hum Mol Genet 2015; 24:6721-35. [PMID: 26362250 PMCID: PMC4634376 DOI: 10.1093/hmg/ddv377] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/27/2015] [Accepted: 09/08/2015] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) and related tauopathies comprise a large group of neurodegenerative diseases associated with the pathological aggregation of tau protein. While much effort has focused on understanding the function of tau, little is known about the endogenous mechanisms regulating tau metabolism in vivo and how these contribute to disease. Previously, we have shown that the microRNA (miRNA) cluster miR-132/212 is downregulated in tauopathies such as AD. Here, we report that miR-132/212 deficiency in mice leads to increased tau expression, phosphorylation and aggregation. Using reporter assays and cell-based studies, we demonstrate that miR-132 directly targets tau mRNA to regulate its expression. We identified GSK-3β and PP2B as effectors of abnormal tau phosphorylation in vivo. Deletion of miR-132/212 induced tau aggregation in mice expressing endogenous or human mutant tau, an effect associated with autophagy dysfunction. Conversely, treatment of AD mice with miR-132 mimics restored in part memory function and tau metabolism. Finally, miR-132 and miR-212 levels correlated with insoluble tau and cognitive impairment in humans. These findings support a role for miR-132/212 in the regulation of tau pathology in mice and humans and provide new alternatives for therapeutic development.
Collapse
Affiliation(s)
- Pascal Y Smith
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Julia Hernandez-Rapp
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Francis Jolivette
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Cynthia Lecours
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Médecine Moléculaire
| | - Kanchan Bisht
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Médecine Moléculaire
| | - Claudia Goupil
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Veronique Dorval
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Sepideh Parsi
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Françoise Morin
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - Emmanuel Planel
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Francisco-Jose Fernandez-Gomez
- Faculté de Médecine, Université de Lille, UDSL, Lille F-59045, France and UMR-S 1172, Alzheimer and Tauopathies, Inserm, Lille F-59045, France
| | - Nicolas Sergeant
- Faculté de Médecine, Université de Lille, UDSL, Lille F-59045, France and UMR-S 1172, Alzheimer and Tauopathies, Inserm, Lille F-59045, France
| | - Luc Buée
- Faculté de Médecine, Université de Lille, UDSL, Lille F-59045, France and UMR-S 1172, Alzheimer and Tauopathies, Inserm, Lille F-59045, France
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Médecine Moléculaire
| | - Frédéric Calon
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Faculté de Pharmacie, Université Laval, Québec, QC, Canada G1V 0A6
| | - Sébastien S Hébert
- Axe Neurosciences, Centre de Recherche du CHU de Québec, CHUL, Québec, QC, Canada G1V 4G2, Département de Psychiatrie et Neurosciences,
| |
Collapse
|
146
|
Heyer MP, Kenny PJ. Corticostriatal microRNAs in addiction. Brain Res 2015; 1628:2-16. [DOI: 10.1016/j.brainres.2015.07.047] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 07/11/2015] [Accepted: 07/25/2015] [Indexed: 01/28/2023]
|
147
|
Weinberg RB, Mufson EJ, Counts SE. Evidence for a neuroprotective microRNA pathway in amnestic mild cognitive impairment. Front Neurosci 2015; 9:430. [PMID: 26594146 PMCID: PMC4633499 DOI: 10.3389/fnins.2015.00430] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/22/2015] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs (miRNAs) that regulate mRNA stability have been linked to amyloid production, tau phosphorylation, and inflammation in Alzheimer's disease (AD). However, whether cerebral miRNA networks are dysregulated during the earliest stages of AD remains underexplored. We performed miRNA expression analysis using frontal cortex tissue harvested from subjects who died with a clinical diagnosis of no cognitive impairment (NCI), amnestic mild cognitive impairment (aMCI, a putative prodromal AD stage), or mild AD. Analysis revealed that the miRNA clusters miR-212/132 and miR-23a/23b were down-regulated in the frontal cortex of aMCI subjects. Both miR-212/132 and miR23a/b are predicted to destabilize the message for sirtuin 1 (sirt1); hence, down-regulation of either miR-212/132 or miR-23a/b in frontal cortex should promote sirt1 mRNA expression in this region. qPCR studies revealed that frontal cortex levels of sirt1 were increased in aMCI. Given the ability of frontal cortex to respond to the onset of dementia by neuronal reorganization, these data suggest that miRNA-mediated up-regulation of the sirt1 pathway represents a compensatory response to the onset of the disease. By contrast, qPCR analysis of inferior temporal cortex, an area affected early in the progression of AD, showed no changes in miR-212/132, miR-23a/b, or sirt1 transcripts in the same aMCI subjects. In vitro mechanistic studies showed that coordinated down-regulation of miR-212 and miR-23a increased sirt1 protein expression and provided neuroprotection from β-amyloid toxicity in human neuronal cells. Taken together, these data suggest a novel miRNA-mediated neuroprotective pathway activated during the progression of AD that may be amenable to therapeutic manipulation.
Collapse
Affiliation(s)
- Rebecca B. Weinberg
- Department of Translational Science and Molecular Medicine, Michigan State UniversityGrand Rapids, MI, USA
| | - Elliott J. Mufson
- Department of Neurobiology, Barrow Neurological InstitutePhoenix, AZ, USA
| | - Scott E. Counts
- Department of Translational Science and Molecular Medicine, Michigan State UniversityGrand Rapids, MI, USA
- Department of Family Medicine, Michigan State UniversityGrand Rapids, MI, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's HospitalGrand Rapids, MI, USA
| |
Collapse
|
148
|
Alsharafi WA, Xiao B, Abuhamed MM, Luo Z. miRNAs: biological and clinical determinants in epilepsy. Front Mol Neurosci 2015; 8:59. [PMID: 26528124 PMCID: PMC4602137 DOI: 10.3389/fnmol.2015.00059] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 09/18/2015] [Indexed: 12/22/2022] Open
Abstract
Recently, microRNAs (miRNAs) are reported to be crucial modulators in the pathogenesis and potential treatment of epilepsies. To date, several miRNAs have been demonstrated to be significantly expressed in the epileptic tissues and strongly associated with the development of epilepsy. Specifically, miRNAs regulate synaptic strength, inflammation, neuronal and glial function, ion channels, and apoptosis. Furthermore, peripheral blood miRNAs can also be utilized as diagnostic biomarkers to assess disease risk and treatment responses. Here, we will summarize the recent available literature regarding the role of miRNAs in the pathogenesis and treatment of epilepsy. Moreover, we will provide brief insight into the potential of miRNA as diagnostic biomarkers for early diagnosis and prognosis of epilepsy.
Collapse
Affiliation(s)
- Walid A Alsharafi
- Department of Neurology, Xiangya Hospital, Central South University Changsha, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University Changsha, China
| | | | - Zhaohui Luo
- Department of Neurology, Xiangya Hospital, Central South University Changsha, China
| |
Collapse
|
149
|
Pan Y, Liu R, Terpstra E, Wang Y, Qiao F, Wang J, Tong Y, Pan B. Dysregulation and Diagnostic Potential of microRNA in Alzheimer’s Disease. J Alzheimers Dis 2015; 49:1-12. [DOI: 10.3233/jad-150451] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Yaoqian Pan
- Department of Veterinary Pathology, College of Animal Sciences, Henan Institute of Science and Technology, Xinxiang, China
| | - Ruizhu Liu
- China-Japan Union Hospital Jilin University, Changchun, Jilin, China
| | - Erin Terpstra
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD, USA
| | - Yanqing Wang
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD, USA
| | - Fangfang Qiao
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD, USA
| | - Jin Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- State Key Lab of Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yigang Tong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Bo Pan
- Department of Veterinary Pathology, College of Animal Sciences, Henan Institute of Science and Technology, Xinxiang, China
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD, USA
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- State Key Lab of Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
150
|
Parsi S, Smith PY, Goupil C, Dorval V, Hébert SS. Preclinical Evaluation of miR-15/107 Family Members as Multifactorial Drug Targets for Alzheimer's Disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e256. [PMID: 26440600 PMCID: PMC4881761 DOI: 10.1038/mtna.2015.33] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 08/20/2015] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial, fatal neurodegenerative disorder characterized by the abnormal accumulation of Aβ and Tau deposits in the brain. There is no cure for AD, and failure at different clinical trials emphasizes the need for new treatments. In recent years, significant progress has been made toward the development of miRNA-based therapeutics for human disorders. This study was designed to evaluate the efficiency and potential safety of miRNA replacement therapy in AD, using miR-15/107 paralogues as candidate drug targets. We identified miR-16 as a potent inhibitor of amyloid precursor protein (APP) and BACE1 expression, Aβ peptide production, and Tau phosphorylation in cells. Brain delivery of miR-16 mimics in mice resulted in a reduction of AD-related genes APP, BACE1, and Tau in a region-dependent manner. We further identified Nicastrin, a γ-secretase component involved in Aβ generation, as a target of miR-16. Proteomics analysis identified a number of additional putative miR-16 targets in vivo, including α-Synuclein and Transferrin receptor 1. Top-ranking biological networks associated with miR-16 delivery included AD and oxidative stress. Collectively, our data suggest that miR-16 is a good candidate for future drug development by targeting simultaneously endogenous regulators of AD biomarkers (i.e., Aβ and Tau), inflammation, and oxidative stress.
Collapse
Affiliation(s)
- Sepideh Parsi
- Centre de recherche du CHU de Québec, CHUL, Axe Neurosciences, Québec, Québec City, Canada.,Département de psychiatrie et neurosciences, Université Laval, Québec, Québec City, Canada
| | - Pascal Y Smith
- Centre de recherche du CHU de Québec, CHUL, Axe Neurosciences, Québec, Québec City, Canada.,Département de psychiatrie et neurosciences, Université Laval, Québec, Québec City, Canada
| | - Claudia Goupil
- Centre de recherche du CHU de Québec, CHUL, Axe Neurosciences, Québec, Québec City, Canada.,Département de psychiatrie et neurosciences, Université Laval, Québec, Québec City, Canada
| | - Véronique Dorval
- Centre de recherche du CHU de Québec, CHUL, Axe Neurosciences, Québec, Québec City, Canada.,Département de psychiatrie et neurosciences, Université Laval, Québec, Québec City, Canada
| | - Sébastien S Hébert
- Centre de recherche du CHU de Québec, CHUL, Axe Neurosciences, Québec, Québec City, Canada.,Département de psychiatrie et neurosciences, Université Laval, Québec, Québec City, Canada
| |
Collapse
|