151
|
Prognostic serum miRNA biomarkers associated with Alzheimer's disease shows concordance with neuropsychological and neuroimaging assessment. Mol Psychiatry 2015; 20:1188-96. [PMID: 25349172 DOI: 10.1038/mp.2014.127] [Citation(s) in RCA: 282] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/10/2014] [Accepted: 08/25/2014] [Indexed: 02/06/2023]
Abstract
There is no consensus for a blood-based test for the early diagnosis of Alzheimer's disease (AD). Expression profiling of small non-coding RNA's, microRNA (miRNA), has revealed diagnostic potential in human diseases. Circulating miRNA are found in small vesicles known as exosomes within biological fluids such as human serum. The aim of this work was to determine a set of differential exosomal miRNA biomarkers between healthy and AD patients, which may aid in diagnosis. Using next-generation deep sequencing, we profiled exosomal miRNA from serum (N=49) collected from the Australian Imaging, Biomarkers and Lifestyle Flagship Study (AIBL). Sequencing results were validated using quantitative reverse transcription PCR (qRT-PCR; N=60), with predictions performed using the Random Forest method. Additional risk factors collected during the 4.5-year AIBL Study including clinical, medical and cognitive assessments, and amyloid neuroimaging with positron emission tomography were assessed. An AD-specific 16-miRNA signature was selected and adding established risk factors including age, sex and apolipoprotein ɛ4 (APOE ɛ4) allele status to the panel of deregulated miRNA resulted in a sensitivity and specificity of 87% and 77%, respectively, for predicting AD. Furthermore, amyloid neuroimaging information for those healthy control subjects incorrectly classified with AD-suggested progression in these participants towards AD. These data suggest that an exosomal miRNA signature may have potential to be developed as a suitable peripheral screening tool for AD.
Collapse
|
152
|
Xiong Y, Liu F, Liu D, Huang H, Wei N, Tan L, Chen J, Man H, Gong C, Lu Y, Wang J, Zhu L. Opposite effects of two estrogen receptors on tau phosphorylation through disparate effects on the miR-218/PTPA pathway. Aging Cell 2015; 14:867-77. [PMID: 26111662 PMCID: PMC4568974 DOI: 10.1111/acel.12366] [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] [Accepted: 05/27/2015] [Indexed: 12/14/2022] Open
Abstract
The two estrogen receptors (ERs), ERα and ERβ, mediate the diverse biological functions of estradiol. Opposite effects of ERα and ERβ have been found in estrogen-induced cancer cell proliferation and differentiation as well as in memory-related tasks. However, whether these opposite effects are implicated in the pathogenesis of Alzheimer’s disease (AD) remains unclear. Here, we find that ERα and ERβ play contrasting roles in regulating tau phosphorylation, which is a pathological hallmark of AD. ERα increases the expression of miR-218 to suppress the protein levels of its specific target, protein tyrosine phosphatase α (PTPα). The downregulation of PTPα results in the abnormal tyrosine hyperphosphorylation of glycogen synthase kinase-3β (resulting in activation) and protein phosphatase 2A (resulting in inactivation), the major tau kinase and phosphatase. Suppressing the increased expression of miR-218 inhibits the ERα-induced tau hyperphosphorylation as well as the PTPα decline. In contrast, ERβ inhibits tau phosphorylation by limiting miR-218 levels and restoring the miR-218 levels antagonized the attenuation of tau phosphorylation by ERβ. These data reveal for the first time opposing roles for ERα and ERβ in AD pathogenesis and suggest potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Yan‐Si Xiong
- Department of Pathophysiology School of Basic Medicine Key Laboratory of Neurological Disorder of Education Ministry Tongji Medical College Huazhong University of Science and Technology Wuhan 430030China
- The Institute for Brain Research Collaborative Innovation Center for Brain Science Huazhong University of Science and Technology Wuhan 430030 China
| | - Fang‐Fang Liu
- Department of Pathophysiology School of Basic Medicine Key Laboratory of Neurological Disorder of Education Ministry Tongji Medical College Huazhong University of Science and Technology Wuhan 430030China
- The Institute for Brain Research Collaborative Innovation Center for Brain Science Huazhong University of Science and Technology Wuhan 430030 China
| | - Dan Liu
- The Institute for Brain Research Collaborative Innovation Center for Brain Science Huazhong University of Science and Technology Wuhan 430030 China
- Department of Genetics School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030China
- Sino‐Canada Collaborative Platform on Molecular Biology of Neurological Disease Tongji Medical College Huazhong University of Science and Technology Wuhan 430030China
| | - He‐Zhou Huang
- Department of Pathophysiology School of Basic Medicine Key Laboratory of Neurological Disorder of Education Ministry Tongji Medical College Huazhong University of Science and Technology Wuhan 430030China
- The Institute for Brain Research Collaborative Innovation Center for Brain Science Huazhong University of Science and Technology Wuhan 430030 China
| | - Na Wei
- Department of Pathophysiology School of Basic Medicine Key Laboratory of Neurological Disorder of Education Ministry Tongji Medical College Huazhong University of Science and Technology Wuhan 430030China
- The Institute for Brain Research Collaborative Innovation Center for Brain Science Huazhong University of Science and Technology Wuhan 430030 China
| | - Lu Tan
- Department of Pathophysiology School of Basic Medicine Key Laboratory of Neurological Disorder of Education Ministry Tongji Medical College Huazhong University of Science and Technology Wuhan 430030China
- The Institute for Brain Research Collaborative Innovation Center for Brain Science Huazhong University of Science and Technology Wuhan 430030 China
| | - Jian‐Guo Chen
- The Institute for Brain Research Collaborative Innovation Center for Brain Science Huazhong University of Science and Technology Wuhan 430030 China
- Department of Pharmacology School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan 430030 China
| | - Heng‐Ye Man
- Department of Biology Boston University Boston MA 02215USA
| | - Cheng‐Xin Gong
- Department of Neurochemistry Inge Grundke‐Iqbal Research Floor New York State Institute for Basic Research in Developmental Disabilities Staten Island NY 10314USA
| | - Youming Lu
- The Institute for Brain Research Collaborative Innovation Center for Brain Science Huazhong University of Science and Technology Wuhan 430030 China
| | - Jian‐Zhi Wang
- Department of Pathophysiology School of Basic Medicine Key Laboratory of Neurological Disorder of Education Ministry Tongji Medical College Huazhong University of Science and Technology Wuhan 430030China
- The Institute for Brain Research Collaborative Innovation Center for Brain Science Huazhong University of Science and Technology Wuhan 430030 China
| | - Ling‐Qiang Zhu
- Department of Pathophysiology School of Basic Medicine Key Laboratory of Neurological Disorder of Education Ministry Tongji Medical College Huazhong University of Science and Technology Wuhan 430030China
- The Institute for Brain Research Collaborative Innovation Center for Brain Science Huazhong University of Science and Technology Wuhan 430030 China
- Sino‐Canada Collaborative Platform on Molecular Biology of Neurological Disease Tongji Medical College Huazhong University of Science and Technology Wuhan 430030China
| |
Collapse
|
153
|
Butler AA, Webb WM, Lubin FD. Regulatory RNAs and control of epigenetic mechanisms: expectations for cognition and cognitive dysfunction. Epigenomics 2015; 8:135-51. [PMID: 26366811 DOI: 10.2217/epi.15.79] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The diverse functions of noncoding RNAs (ncRNAs) can influence virtually every aspect of the transcriptional process including epigenetic regulation of genes. In the CNS, regulatory RNA networks and epigenetic mechanisms have broad relevance to gene transcription changes involved in long-term memory formation and cognition. Thus, it is becoming increasingly clear that multiple classes of ncRNAs impact neuronal development, neuroplasticity, and cognition. Currently, a large gap exists in our knowledge of how ncRNAs facilitate epigenetic processes, and how this phenomenon affects cognitive function. In this review, we discuss recent findings highlighting a provocative role for ncRNAs including lncRNAs and piRNAs in the control of epigenetic mechanisms involved in cognitive function. Furthermore, we discuss the putative roles for these ncRNAs in cognitive disorders such as schizophrenia and Alzheimer's disease.
Collapse
Affiliation(s)
- Anderson A Butler
- Department of Neurobiology, University of Alabama at Birmingham, 1825 University Boulevard, Birmingham, AL 35294, USA
| | - William M Webb
- Department of Neurobiology, University of Alabama at Birmingham, 1825 University Boulevard, Birmingham, AL 35294, USA
| | - Farah D Lubin
- Department of Neurobiology, University of Alabama at Birmingham, 1825 University Boulevard, Birmingham, AL 35294, USA
| |
Collapse
|
154
|
Gupta S, Verma S, Mantri S, Berman NE, Sandhir R. Targeting MicroRNAs in Prevention and Treatment of Neurodegenerative Disorders. Drug Dev Res 2015; 76:397-418. [PMID: 26359796 DOI: 10.1002/ddr.21277] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Preclinical Research microRNAs (miRNAs) are small noncoding RNAs (ncRNAs) that are key regulators of gene expression. They act on wide range of targets by binding to mRNA via imperfect complementarity at 3' UTR. Evidence suggests that miRNAs regulate many biological processes including neuronal development, differentiation, and disease. Altered expression of several miRNAs has been reported in many neurodegenerative disorders (NDDs). Many miRNAs are altered in these diseases, but miRNA 15, miRNA 21, and miRNA 146a have been shown to play critical role in many neurodegenerative conditions. As these miRNAs regulate many genes, miRNA targeted approaches would allow concurrently targeting of multiple effectors of pathways that regulate disease progression. In this review, we describe the role of miRNAs in various NDDs and their potential as therapeutic tools in prevention and treatment of neurological conditions.
Collapse
Affiliation(s)
- Smriti Gupta
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Savita Verma
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Shrikant Mantri
- Computational Biology Laboratory, National Agri-Food Biotechnology Institute, Mohali, Punjab, 160071, India
| | - Nancy E Berman
- Department of Anatomy & Cell Biology, Kansas University Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160, USA
| | - Rajat Sandhir
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| |
Collapse
|
155
|
Lardenoije R, Iatrou A, Kenis G, Kompotis K, Steinbusch HWM, Mastroeni D, Coleman P, Lemere CA, Hof PR, van den Hove DLA, Rutten BPF. The epigenetics of aging and neurodegeneration. Prog Neurobiol 2015; 131:21-64. [PMID: 26072273 PMCID: PMC6477921 DOI: 10.1016/j.pneurobio.2015.05.002] [Citation(s) in RCA: 246] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 05/13/2015] [Accepted: 05/13/2015] [Indexed: 12/14/2022]
Abstract
Epigenetics is a quickly growing field encompassing mechanisms regulating gene expression that do not involve changes in the genotype. Epigenetics is of increasing relevance to neuroscience, with epigenetic mechanisms being implicated in brain development and neuronal differentiation, as well as in more dynamic processes related to cognition. Epigenetic regulation covers multiple levels of gene expression; from direct modifications of the DNA and histone tails, regulating the level of transcription, to interactions with messenger RNAs, regulating the level of translation. Importantly, epigenetic dysregulation currently garners much attention as a pivotal player in aging and age-related neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, where it may mediate interactions between genetic and environmental risk factors, or directly interact with disease-specific pathological factors. We review current knowledge about the major epigenetic mechanisms, including DNA methylation and DNA demethylation, chromatin remodeling and non-coding RNAs, as well as the involvement of these mechanisms in normal aging and in the pathophysiology of the most common neurodegenerative diseases. Additionally, we examine the current state of epigenetics-based therapeutic strategies for these diseases, which either aim to restore the epigenetic homeostasis or skew it to a favorable direction to counter disease pathology. Finally, methodological challenges of epigenetic investigations and future perspectives are discussed.
Collapse
Affiliation(s)
- Roy Lardenoije
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands
| | - Artemis Iatrou
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands
| | - Gunter Kenis
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands
| | - Konstantinos Kompotis
- Center for Integrative Genomics, University of Lausanne, Genopode Building, 1015 Lausanne-Dorigny, Switzerland
| | - Harry W M Steinbusch
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands
| | - Diego Mastroeni
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands; L.J. Roberts Alzheimer's Disease Center, Banner Sun Health Research Institute, 10515 W. Santa Fe Drive, Sun City, AZ 85351, USA
| | - Paul Coleman
- L.J. Roberts Alzheimer's Disease Center, Banner Sun Health Research Institute, 10515 W. Santa Fe Drive, Sun City, AZ 85351, USA
| | - Cynthia A Lemere
- Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Daniel L A van den Hove
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands; Laboratory of Translational Neuroscience, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Fuechsleinstrasse 15, 97080 Wuerzburg, Germany
| | - Bart P F Rutten
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands.
| |
Collapse
|
156
|
Bajic V, Spremo-Potparevic B, Zivkovic L, Isenovic ER, Arendt T. Cohesion and the aneuploid phenotype in Alzheimer's disease: A tale of genome instability. Neurosci Biobehav Rev 2015; 55:365-74. [PMID: 26003528 DOI: 10.1016/j.neubiorev.2015.05.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 03/26/2015] [Accepted: 05/12/2015] [Indexed: 12/20/2022]
Abstract
Neurons are postmitotic cells that are in permanent cell cycle arrest. However, components of the cell cycle machinery that are expressed in Alzheimer's disease (AD) neurons are showing features of a cycling cell and those attributed to a postmitotic cell as well. Furthermore, the unique physiological operations taking place in neurons, ascribed to "core cell cycle regulators" are also key regulators in cell division. Functions of these cell cycle regulators include neuronal migration, axonal elongation, axon pruning, dendrite morphogenesis and synaptic maturation and plasticity. In this review, we focus on cohesion and cohesion related proteins in reference to their neuronal functions and how impaired centromere/cohesion dynamics may connect cell cycle dysfunction to aneuploidy in AD.
Collapse
Affiliation(s)
- Vladan Bajic
- Institute for Nuclear Research "Vinca", Laboratory for Radiobiology and Molecular Genetics, University of Belgrade, P.O. 522, 11001 Belgrade, Serbia.
| | - Biljana Spremo-Potparevic
- Faculty of Pharmacy, Institute of Physiology, Department of Biology and Human Genetics, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia.
| | - Lada Zivkovic
- Faculty of Pharmacy, Institute of Physiology, Department of Biology and Human Genetics, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia.
| | - Esma R Isenovic
- Institute for Nuclear Research "Vinca", Laboratory for Radiobiology and Molecular Genetics, University of Belgrade, P.O. 522, 11001 Belgrade, Serbia.
| | - Thomas Arendt
- Paul-Flechsig-Institute for Brain Research, University of Leipzig, School of Medicine, Leipzig, Germany.
| |
Collapse
|
157
|
Abstract
MicroRNAs (miRNAs) are key players in the regulation of neuronal processes by targeting a large network of target messenger RNAs (mRNAs). However, the identity and function of mRNAs targeted by miRNAs in specific cells of the brain are largely unknown. Here, we established an adeno-associated viral vector (AAV)-based neuron-specific Argonaute2:GFP-RNA immunoprecipitation followed by high-throughput sequencing to analyse the regulatory role of miRNAs in mouse hippocampal neurons. Using this approach, we identified more than two thousand miRNA targets in hippocampal neurons, regulating essential neuronal features such as cell signalling, transcription and axon guidance. Furthermore, we found that stable inhibition of the highly expressed miR-124 and miR-125 in hippocampal neurons led to significant but distinct changes in the AGO2 binding of target mRNAs, resulting in subsequent upregulation of numerous miRNA target genes. These findings greatly enhance our understanding of the miRNA targetome in hippocampal neurons.
Collapse
|
158
|
Rodriguez-Ortiz CJ, Baglietto-Vargas D, Martinez-Coria H, LaFerla FM, Kitazawa M. Upregulation of miR-181 decreases c-Fos and SIRT-1 in the hippocampus of 3xTg-AD mice. J Alzheimers Dis 2015; 42:1229-38. [PMID: 25024332 DOI: 10.3233/jad-140204] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
MicroRNAs are a group of small RNAs that regulate diverse cellular processes including neuronal function. Recent studies have shown that dysregulation of specific microRNAs is critically involved in the development of Alzheimer's disease (AD). Most of these reports have focused on microRNAs implicated in alterations of amyloid-β and tau. However, studies exploring the relation between microRNAs dysregulation in AD and synaptic plasticity are scarce despite the well-known involvement of microRNAs in synaptic plasticity. Since impairments in synaptic plasticity and neuronal loss are two important features displayed in AD patients, it is feasible to hypothesize that alterations in plasticity-related microRNAs underlie AD progression. Here, levels of a small number of microRNAs implicated in normal neuronal function and/or plasticity were examined in an AD model. Twelve-month old 3xTg-AD mice with plaques and tangles presented a significant upregulation of miR-181 in the hippocampus compared to age-matched wild type mice. Increased miR-181 was not detected in pre-pathological 3xTg-AD mice. Analysis of predicted targets of miR-181 identified c-Fos and SIRT-1, proteins critically involved in memory formation. Both c-Fos and SIRT-1 levels were significantly decreased in the ventral hippocampus of twelve-month old 3xTg-AD mice. Overexpression of miR-181 in SH-SY5Y cells significantly decreased c-Fos and SIRT-1, strongly suggesting that miR-181 directly regulates the expression of these two proteins. These findings indicate a connection between miR-181 and proteins involve in synaptic plasticity and memory processing in a transgenic mouse model of AD. Our results suggest that microRNAs involved in synaptic plasticity might be an important factor that contributes to AD neuropathology.
Collapse
Affiliation(s)
| | - David Baglietto-Vargas
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA Institute for Memory Impairments and Neurological Diseases, University of California, Irvine, CA, USA
| | - Hilda Martinez-Coria
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA Institute for Memory Impairments and Neurological Diseases, University of California, Irvine, CA, USA
| | - Frank M LaFerla
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA Institute for Memory Impairments and Neurological Diseases, University of California, Irvine, CA, USA
| | - Masashi Kitazawa
- School of Natural Sciences, University of California, Merced, CA, USA
| |
Collapse
|
159
|
Millan MJ. The epigenetic dimension of Alzheimer's disease: causal, consequence, or curiosity? DIALOGUES IN CLINICAL NEUROSCIENCE 2015. [PMID: 25364287 PMCID: PMC4214179 DOI: 10.31887/dcns.2014.16.3/mmillan] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Early-onset, familial Alzheimer's disease (AD) is rare and may be attributed to disease-causinq mutations. By contrast, late onset, sporadic (non-Mendelian) AD is far more prevalent and reflects the interaction of multiple genetic and environmental risk factors, together with the disruption of epigenetic mechanisms controlling gene expression. Accordingly, abnormal patterns of histone acetylation and methylation, as well as anomalies in global and promoter-specific DNA methylation, have been documented in AD patients, together with a deregulation of noncoding RNA. In transgenic mouse models for AD, epigenetic dysfunction is likewise apparent in cerebral tissue, and it has been directly linked to cognitive and behavioral deficits in functional studies. Importantly, epigenetic deregulation interfaces with core pathophysiological processes underlying AD: excess production of Aβ42, aberrant post-translational modification of tau, deficient neurotoxic protein clearance, axonal-synaptic dysfunction, mitochondrial-dependent apoptosis, and cell cycle re-entry. Reciprocally, DNA methylation, histone marks and the levels of diverse species of microRNA are modulated by Aβ42, oxidative stress and neuroinflammation. In conclusion, epigenetic mechanisms are broadly deregulated in AD mainly upstream, but also downstream, of key pathophysiological processes. While some epigenetic shifts oppose the evolution of AD, most appear to drive its progression. Epigenetic changes are of irrefutable importance for AD, but they await further elucidation from the perspectives of pathogenesis, biomarkers and potential treatment.
Collapse
Affiliation(s)
- Mark J Millan
- Pole of Innovation in Neuropsychiatry, Institut de Recherche Servier, Croissy-sur-Seine, France
| |
Collapse
|
160
|
Minocycline reduces neuroinflammation but does not ameliorate neuron loss in a mouse model of neurodegeneration. Sci Rep 2015; 5:10535. [PMID: 26000566 PMCID: PMC4441131 DOI: 10.1038/srep10535] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 04/23/2015] [Indexed: 12/13/2022] Open
Abstract
Minocycline is a broad-spectrum tetracycline antibiotic. A number of preclinical studies have shown that minocycline exhibits neuroprotective effects in various animal models of neurological diseases. However, it remained unknown whether minocycline is effective to prevent neuron loss. To systematically evaluate its effects, minocycline was used to treat Dicer conditional knockout (cKO) mice which display age-related neuron loss. The drug was given to mutant mice prior to the occurrence of neuroinflammation and neurodegeneration, and the treatment had lasted 2 months. Levels of inflammation markers, including glial fibrillary acidic protein (GFAP), ionized calcium-binding adapter molecule1 (Iba1) and interleukin6 (IL6), were significantly reduced in minocycline-treated Dicer cKO mice. In contrast, levels of neuronal markers and the total number of apoptotic cells in Dicer cKO mice were not affected by the drug. In summary, inhibition of neuroinflammation by minocycline is insufficient to prevent neuron loss and apoptosis.
Collapse
|
161
|
Abstract
Recent studies have revealed that patients with psychiatric disorders have altered microRNA (miRNA) expression profiles in the circulation and brain. Furthermore, animal studies have shown that manipulating the levels of particular miRNAs in the brain can alter behaviour. Here, we review recent studies in humans, animal models, cellular systems and bioinformatics that have advanced our understanding of the contribution of brain miRNAs to the regulation of behaviour in the context of psychiatric conditions. These studies highlight the potential of miRNA levels to be used in the diagnosis of psychiatric disorders and suggest that brain miRNAs could become novel treatment targets for psychiatric disorders.
Collapse
|
162
|
Are microRNAs the Molecular Link Between Metabolic Syndrome and Alzheimer's Disease? Mol Neurobiol 2015; 53:2320-38. [PMID: 25976367 DOI: 10.1007/s12035-015-9201-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/29/2015] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in people over 65 years of age. At present, treatment options for AD address only its symptoms, and there are no available treatments for the prevention or delay of the disease process. Several preclinical and epidemiological studies have linked metabolic risk factors such as hypertension, obesity, dyslipidemia, and diabetes to the pathogenesis of AD. However, the molecular mechanisms that underlie this relationship are not fully understood. Considering that less than 1% of cases of AD are attributable to genetic factors, the identification of new molecular targets linking metabolic risk factors to neuropathological processes is necessary for improving the diagnosis and treatment of AD. The dysregulation of microRNAs (miRNAs), small non-coding RNAs that regulate several biological processes, has been implicated in the development of different pathologies. In this review, we summarize some of the relevant evidence that points to the role of miRNAs in metabolic syndrome (MetS) and AD and propose that miRNAs may be a molecular link in the complex relationship between both diseases.
Collapse
|
163
|
Arisi I, D'Onofrio M, Brandi R, Cattaneo A, Bertolazzi P, Cumbo F, Felici G, Guerra C. Time dynamics of protein complexes in the AD11 transgenic mouse model for Alzheimer's disease like pathology. BMC Neurosci 2015; 16:28. [PMID: 25925689 PMCID: PMC4436769 DOI: 10.1186/s12868-015-0155-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/11/2015] [Indexed: 11/20/2022] Open
Abstract
Background Many approaches exist to integrate protein-protein interaction data with other sources of information, most notably with gene co-expression data, to obtain information on network dynamics. It is of interest to look at groups of interacting gene products that form a protein complex. We were interested in applying new tools to the characterization of pathogenesis and dynamic events of an Alzheimer’s-like neurodegenerative model, the AD11 mice, expressing an anti-NGF monoclonal antibody. The goal was to quantify the impact of neurodegeneration on protein complexes, by measuring the correlation between gene expression data by different metrics. Results Data were extracted from the gene expression profile of AD11 brain, obtained by Agilent microarray, at 1, 3, 6, 15 months of age. For genes coding proteins in complexes, the correlation matrix of pairwise expression was computed. The dynamics between correlation matrices at different time points was evaluated: paired T-test between average correlation levels and a normalized Euclidean distance with z-score. We unveiled a differential wiring of interactions in a set of complexes, whose network structure discriminates between transgenic and control mice. Furthermore, we analyzed the dynamics of gene expression values, by looking at changes in gene-to-gene correlation over time and identified those complexes that exhibit a different timedependent behaviour between transgenic and controls. The most significant changes in correlation dynamics are concentrated in the early stage of disease, with higher correlation in AD11 mice compared to controls. Many complexes go through dynamic changes over time, showing the role of the dysfunctional immunoproteasome, as early neurodegenerative disease event. Furthermore, this analysis shows key events in the neurodegeneration process of the AD11 model, by identifying significant differences in co-expression values of other complexes, such as parvulin complex, with a role in protein misfolding and proteostasis, and of complexes involved in transcriptional mechanisms. Conclusions We have proposed a novel approach to analyze the network structure of protein complexes, by two different measures to evaluate the dynamics of gene-gene correlation matrices from gene expression profiles. The methodology was able to investigate the re-organization of interactions within protein complexes in the AD11 model of neurodegeneration.
Collapse
Affiliation(s)
- Ivan Arisi
- Genomics Facility, European Brain Research Institute (EBRI) Rita Levi-Montalcini, Via del Fosso di Fiorano, 64, 00143, Rome, Italy.
| | - Mara D'Onofrio
- Genomics Facility, European Brain Research Institute (EBRI) Rita Levi-Montalcini, Via del Fosso di Fiorano, 64, 00143, Rome, Italy.
| | - Rossella Brandi
- Genomics Facility, European Brain Research Institute (EBRI) Rita Levi-Montalcini, Via del Fosso di Fiorano, 64, 00143, Rome, Italy.
| | - Antonino Cattaneo
- Neurotrophic Factors and Neurodegenerative Diseases Unit, EBRI, Rome, Italy. .,Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126, Pisa, Italy.
| | - Paola Bertolazzi
- Istituto di Analisi dei Sistemi ed Informatica "Antonio Ruberti" (IASI-CNR), Rome, Italy.
| | - Fabio Cumbo
- Istituto di Analisi dei Sistemi ed Informatica "Antonio Ruberti" (IASI-CNR), Rome, Italy.
| | - Giovanni Felici
- Istituto di Analisi dei Sistemi ed Informatica "Antonio Ruberti" (IASI-CNR), Rome, Italy.
| | - Concettina Guerra
- Istituto di Analisi dei Sistemi ed Informatica "Antonio Ruberti" (IASI-CNR), Rome, Italy. .,College of Computing, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
164
|
Long noncoding RNA SPRY4-IT1 predicts poor patient prognosis and promotes tumorigenesis in gastric cancer. Tumour Biol 2015; 53:2016-2028. [PMID: 25835973 DOI: 10.1007/s12035-015-9142-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 03/12/2015] [Indexed: 12/16/2022] Open
Abstract
Gastric cancer (GC) is the second common cause of cancer-related death worldwide. Long noncoding RNAs (lncRNAs) are emerging as novel regulators in the cancer paradigm. However, investigation of lncRNAs on GC is still in its infancy. In this study, we focused on lncRNA SPRY4 intronic transcript 1 (SPRY4-IT1) and investigated its expression pattern, clinical significance, biological function, and molecular mechanism in GC. SPRY4-IT1 expression was examined, and its correlation with clinicopathological characteristics and patient prognosis was analyzed. A series of assays were performed to understand the role of SPRY4-IT1 in GC. SPRY4-IT1 expression was elevated in GC tissues and cell lines, and SPRY4-IT1 levels were highly positively correlated with tumor size, invasion depth, distant metastasis, TNM stage, and reduced overall survival (OS) and disease-free survival (DFS). A multivariate analysis showed that SPRY4-IT1 expression is an independent prognostic factor of OS and DFS in patients with GC. Additionally, the results of in vitro assays showed that the suppression of SPRY4-IT1 expression in GC cell line MKN-45 significantly reduced cell proliferation, colony formation, and cell migration/invasion. Moreover, the tumorigenic effects of SPRY4-IT1 were partially mediated by the regulation of certain cyclins and matrix metalloproteinases (MMPs)-related genes. Our data suggest that SPRY4-IT1 plays a critical role in GC tumorigenesis and may represent a novel prognostic marker and potential therapeutic target in patients with GC.
Collapse
|
165
|
Burns TC, Li MD, Mehta S, Awad AJ, Morgan AA. Mouse models rarely mimic the transcriptome of human neurodegenerative diseases: A systematic bioinformatics-based critique of preclinical models. Eur J Pharmacol 2015; 759:101-17. [PMID: 25814260 DOI: 10.1016/j.ejphar.2015.03.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 03/12/2015] [Accepted: 03/12/2015] [Indexed: 12/12/2022]
Abstract
Translational research for neurodegenerative disease depends intimately upon animal models. Unfortunately, promising therapies developed using mouse models mostly fail in clinical trials, highlighting uncertainty about how well mouse models mimic human neurodegenerative disease at the molecular level. We compared the transcriptional signature of neurodegeneration in mouse models of Alzheimer׳s disease (AD), Parkinson׳s disease (PD), Huntington׳s disease (HD) and amyotrophic lateral sclerosis (ALS) to human disease. In contrast to aging, which demonstrated a conserved transcriptome between humans and mice, only 3 of 19 animal models showed significant enrichment for gene sets comprising the most dysregulated up- and down-regulated human genes. Spearman׳s correlation analysis revealed even healthy human aging to be more closely related to human neurodegeneration than any mouse model of AD, PD, ALS or HD. Remarkably, mouse models frequently upregulated stress response genes that were consistently downregulated in human diseases. Among potential alternate models of neurodegeneration, mouse prion disease outperformed all other disease-specific models. Even among the best available animal models, conserved differences between mouse and human transcriptomes were found across multiple animal model versus human disease comparisons, surprisingly, even including aging. Relative to mouse models, mouse disease signatures demonstrated consistent trends toward preserved mitochondrial function protein catabolism, DNA repair responses, and chromatin maintenance. These findings suggest a more complex and multifactorial pathophysiology in human neurodegeneration than is captured through standard animal models, and suggest that even among conserved physiological processes such as aging, mice are less prone to exhibit neurodegeneration-like changes. This work may help explain the poor track record of mouse-based translational therapies for neurodegeneration and provides a path forward to critically evaluate and improve animal models of human disease.
Collapse
Affiliation(s)
- Terry C Burns
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA.
| | - Matthew D Li
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Swapnil Mehta
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Ahmed J Awad
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Alexander A Morgan
- Department of Neurosurgery, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA
| |
Collapse
|
166
|
Fiorenza A, Lopez-Atalaya JP, Rovira V, Scandaglia M, Geijo-Barrientos E, Barco A. Blocking miRNA Biogenesis in Adult Forebrain Neurons Enhances Seizure Susceptibility, Fear Memory, and Food Intake by Increasing Neuronal Responsiveness. Cereb Cortex 2015; 26:1619-1633. [PMID: 25595182 DOI: 10.1093/cercor/bhu332] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The RNase Dicer is essential for the maturation of most microRNAs, a molecular system that plays an essential role in fine-tuning gene expression. To gain molecular insight into the role of Dicer and the microRNA system in brain function, we conducted 2 complementary RNA-seq screens in the hippocampus of inducible forebrain-restricted Dicer1 mutants aimed at identifying the microRNAs primarily affected by Dicer loss and their targets, respectively. Functional genomics analyses predicted the main biological processes and phenotypes associated with impaired microRNA maturation, including categories related to microRNA biology, signal transduction, seizures, and synaptic transmission and plasticity. Consistent with these predictions, we found that, soon after recombination, Dicer-deficient mice exhibited an exaggerated seizure response, enhanced induction of immediate early genes in response to different stimuli, stronger and more stable fear memory, hyperphagia, and increased excitability of CA1 pyramidal neurons. In the long term, we also observed slow and progressive excitotoxic neurodegeneration. Overall, our results indicate that interfering with microRNA biogenesis causes an increase in neuronal responsiveness and disrupts homeostatic mechanisms that protect the neuron against overactivation, which may explain both the initial and late phenotypes associated with the loss of Dicer in excitatory neurons.
Collapse
Affiliation(s)
- Anna Fiorenza
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n., 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Jose P Lopez-Atalaya
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n., 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Victor Rovira
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n., 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Marilyn Scandaglia
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n., 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Emilio Geijo-Barrientos
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n., 03550 Sant Joan d'Alacant, Alicante, Spain
| | - Angel Barco
- Instituto de Neurociencias (Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n., 03550 Sant Joan d'Alacant, Alicante, Spain
| |
Collapse
|
167
|
Qiu L, Tan EK, Zeng L. microRNAs and Neurodegenerative Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 888:85-105. [PMID: 26663180 DOI: 10.1007/978-3-319-22671-2_6] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
microRNAs (miRNAs) are small, noncoding RNA molecules that through imperfect base-pairing with complementary sequences of target mRNA molecules, typically cleave target mRNA, causing subsequent degradation or translation inhibition. Although an increasing number of studies have identified misregulated miRNAs in the neurodegenerative diseases (NDDs) Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, which suggests that alterations in the miRNA regulatory pathway could contribute to disease pathogenesis, the molecular mechanisms underlying the pathological implications of misregulated miRNA expression and the regulation of the key genes involved in NDDs remain largely unknown. In this chapter, we provide evidence of the function and regulation of miRNAs and their association with the neurological events in NDDs. This will help improve our understanding of how miRNAs govern the biological functions of key pathogenic genes in these diseases, which potentially regulate several pathways involved in the progression of neurodegeneration. Additionally, given the growing interest in the therapeutic potential of miRNAs, we discuss current clinical challenges to developing miRNA-based therapeutics for NDDs.
Collapse
Affiliation(s)
- Lifeng Qiu
- Neural Stem Cell Research Lab, Department of Research, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute, SGH Campus, Singapore, 169856, Singapore
- Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
- Neuroscience and Behavioral Disorders program, Duke-National University of Singapore, Graduate Medical School, Singapore, 169857, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Department of Research, National Neuroscience Institute, Singapore, 308433, Singapore.
- Department of Research, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore.
- Neuroscience and Behavioral Disorders program, Duke-National University of Singapore, Graduate Medical School, Singapore, 169857, Singapore.
| |
Collapse
|
168
|
Pandey A, Jauhari A, Singh T, Singh P, Singh N, Srivastava AK, Khan F, Pant AB, Parmar D, Yadav S. Transactivation of P53 by cypermethrin induced miR-200 and apoptosis in neuronal cells. Toxicol Res (Camb) 2015. [DOI: 10.1039/c5tx00200a] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cypermethrin, a pyrethroid pesticide, has been shown to induce neurotoxicity in adult mammals. However, studies are also needed to explore its toxicity in developing brains and understand its mechanism of action in neurons.
Collapse
Affiliation(s)
- Ankita Pandey
- CSIR-Indian Institute of Toxicology Research
- Lucknow-226001
- India
- Department of Biochemistry
- Jamia Hamdard University
| | - Abhishek Jauhari
- CSIR-Indian Institute of Toxicology Research
- Lucknow-226001
- India
- Academy of Scientific and Innovative Research (AcSIR)
- New Delhi
| | - Tanisha Singh
- CSIR-Indian Institute of Toxicology Research
- Lucknow-226001
- India
| | - Parul Singh
- CSIR-Indian Institute of Toxicology Research
- Lucknow-226001
- India
| | - Nishant Singh
- CSIR-Indian Institute of Toxicology Research
- Lucknow-226001
- India
| | - Ankur Kumar Srivastava
- CSIR-Indian Institute of Toxicology Research
- Lucknow-226001
- India
- Academy of Scientific and Innovative Research (AcSIR)
- New Delhi
| | - Farah Khan
- Department of Biochemistry
- Jamia Hamdard University
- New Delhi-110062
- India
| | | | - Devendra Parmar
- CSIR-Indian Institute of Toxicology Research
- Lucknow-226001
- India
| | - Sanjay Yadav
- CSIR-Indian Institute of Toxicology Research
- Lucknow-226001
- India
| |
Collapse
|
169
|
Wakabayashi K, Mori F, Kakita A, Takahashi H, Utsumi J, Sasaki H. Analysis of microRNA from archived formalin-fixed paraffin-embedded specimens of amyotrophic lateral sclerosis. Acta Neuropathol Commun 2014; 2:173. [PMID: 25497327 PMCID: PMC4279903 DOI: 10.1186/s40478-014-0173-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 12/01/2014] [Indexed: 12/13/2022] Open
Abstract
Background MicroRNAs (miRNAs) are noncoding small RNAs that regulate gene expression. This study investigated whether formalin-fixed paraffin-embedded (FFPE) specimens from postmortem cases of neurodegenerative disorders would be suitable for miRNA profiling. Results Ten FFPE samples from 6 cases of amyotrophic lateral sclerosis (ALS) and 4 neurologically normal controls were selected for miRNA analysis on the basis of the following criteria for RNA quality: (i) a postmortem interval of less than 6 hours, (ii) a formalin fixation time of less than 4 weeks, (iii) an RNA yield per sample of more than 500 ng, and (iv) sufficient quality of the RNA agarose gel image. An overall RNA extraction success rate was 46.2%. For ALS, a total of 364 miRNAs were identified in the motor cortex, 91 being up-regulated and 233 down-regulated. Target genes were predicted using miRNA bioinformatics software, and the data applied to ontology analysis. This indicated that one of the miRNAs up-regulated in ALS (miR-338-3p) had already been identified in leukocytes, serum, cerebrospinal fluid and frozen spinal cord from ALS patients. Conclusion Although analysis was possible for just under half of the specimens examined, we were able to show that informative miRNA data can be derived from archived FFPE samples from postmortem cases of neurodegenerative disorders. Electronic supplementary material The online version of this article (doi:10.1186/s40478-014-0173-z) contains supplementary material, which is available to authorized users.
Collapse
|
170
|
Gstir R, Schafferer S, Scheideler M, Misslinger M, Griehl M, Daschil N, Humpel C, Obermair GJ, Schmuckermair C, Striessnig J, Flucher BE, Hüttenhofer A. Generation of a neuro-specific microarray reveals novel differentially expressed noncoding RNAs in mouse models for neurodegenerative diseases. RNA (NEW YORK, N.Y.) 2014; 20:1929-43. [PMID: 25344396 PMCID: PMC4238357 DOI: 10.1261/rna.047225.114] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 08/27/2014] [Indexed: 05/24/2023]
Abstract
We have generated a novel, neuro-specific ncRNA microarray, covering 1472 ncRNA species, to investigate their expression in different mouse models for central nervous system diseases. Thereby, we analyzed ncRNA expression in two mouse models with impaired calcium channel activity, implicated in Epilepsy or Parkinson's disease, respectively, as well as in a mouse model mimicking pathophysiological aspects of Alzheimer's disease. We identified well over a hundred differentially expressed ncRNAs, either from known classes of ncRNAs, such as miRNAs or snoRNAs or which represented entirely novel ncRNA species. Several differentially expressed ncRNAs in the calcium channel mouse models were assigned as miRNAs and target genes involved in calcium signaling, thus suggesting feedback regulation of miRNAs by calcium signaling. In the Alzheimer mouse model, we identified two snoRNAs, whose expression was deregulated prior to amyloid plaque formation. Interestingly, the presence of snoRNAs could be detected in cerebral spine fluid samples in humans, thus potentially serving as early diagnostic markers for Alzheimer's disease. In addition to known ncRNAs species, we also identified 63 differentially expressed, entirely novel ncRNA candidates, located in intronic or intergenic regions of the mouse genome, genomic locations, which previously have been shown to harbor the majority of functional ncRNAs.
Collapse
Affiliation(s)
- Ronald Gstir
- Division of Genomics and RNomics, Innsbruck Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Simon Schafferer
- Division of Genomics and RNomics, Innsbruck Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Marcel Scheideler
- RNA Biology Group, Institute for Genomics and Bioinformatics, Graz University of Technology, 8010 Graz, Austria
| | - Matthias Misslinger
- Division of Genomics and RNomics, Innsbruck Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Matthias Griehl
- Division of Genomics and RNomics, Innsbruck Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Nina Daschil
- Department of Psychiatry and Psychotherapy, University Clinic of General and Social Psychiatry, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Christian Humpel
- Department of Psychiatry and Psychotherapy, University Clinic of General and Social Psychiatry, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Gerald J Obermair
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Claudia Schmuckermair
- Pharmacology and Toxicology, Institute of Pharmacy, and Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Joerg Striessnig
- Pharmacology and Toxicology, Institute of Pharmacy, and Center for Molecular Biosciences, University of Innsbruck, 6020 Innsbruck, Austria
| | - Bernhard E Flucher
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, 6020 Innsbruck, Austria
| | - Alexander Hüttenhofer
- Division of Genomics and RNomics, Innsbruck Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
171
|
Cheng S, Zhang C, Xu C, Wang L, Zou X, Chen G. Age-dependent neuron loss is associated with impaired adult neurogenesis in forebrain neuron-specific Dicer conditional knockout mice. Int J Biochem Cell Biol 2014; 57:186-96. [DOI: 10.1016/j.biocel.2014.10.029] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 10/09/2014] [Accepted: 10/17/2014] [Indexed: 11/24/2022]
|
172
|
Cloutier F, Marrero A, O'Connell C, Morin P. MicroRNAs as potential circulating biomarkers for amyotrophic lateral sclerosis. J Mol Neurosci 2014; 56:102-12. [PMID: 25433762 DOI: 10.1007/s12031-014-0471-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 11/17/2014] [Indexed: 01/04/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a condition primarily characterized by the selective loss of upper and lower motor neurons. Motor neuron loss gives rise to muscle tissue malfunctions, including weakness, spasticity, atrophy, and ultimately paralysis, with death typically due to respiratory failure within 2 to 5 years of symptoms' onset. The mean delay in time from presentation to diagnosis remains at over 1 year. Biomarkers are urgently needed to facilitate ALS diagnosis and prognosis as well as to act as indicators of therapeutic response in clinical trials. MicroRNAs (miRNAs) are small molecules that can influence posttranscriptional gene expression of a variety of transcript targets. Interestingly, miRNAs can be released into the circulation by pathologically affected tissues. This review presents therapeutic and diagnostic challenges associated with ALS, highlights the potential role of miRNAs in ALS, and discusses the diagnostic potential of these molecules in identifying ALS-specific miRNAs or in distinguishing between the various genotypic and phenotypic forms of ALS.
Collapse
Affiliation(s)
- Frank Cloutier
- Institut de l'Atlantique en Neurosciences Atlantic Institute, Vitalité Health Network, Centre Hospitalier Universitaire Dr Georges-L.-Dumont/Dr. Georges-L.-Dumont University Hospital Centre, Moncton, NB, Canada,
| | | | | | | |
Collapse
|
173
|
Qiu L, Zhang W, Tan EK, Zeng L. Deciphering the function and regulation of microRNAs in Alzheimer's disease and Parkinson's disease. ACS Chem Neurosci 2014; 5:884-94. [PMID: 25210999 DOI: 10.1021/cn500149w] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are single stranded, noncoding RNA molecules that are encoded by eukaryotic nuclear DNA. miRNAs function through imperfect base-pairing with complementary sequences of target mRNA molecules, which is typically via the cleavage of target mRNA with transcriptional repression or translational degradation. An increasing number of studies identified dysregulation of miRNAs in neurodegenerative disease and suggest that alterations in the miRNA regulatory pathway could contribute to the disease pathogenesis. However, molecular mechanisms underlying the pathological implications of dysregulated miRNA expression and regulation of the key genes that are involved in neurodegenerative diseases remain largely unknown. Here, we review the evidence for the functional role of dysregulated miRNAs involved in disease pathogenesis, as well as how miRNAs govern neuronal functions either upstream or downstream of target genes that are disease pathogenic factors. Furthermore, we review the cellular feedback regulation between miRNAs and target genes in neurodegenerative diseases, with a focus on Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Lifeng Qiu
- Neural
Stem Cell Research Lab, Research Department, National Neuroscience Institute, 308433, Singapore
| | - Wei Zhang
- Neural
Stem Cell Research Lab, Research Department, National Neuroscience Institute, 308433, Singapore
| | - Eng King Tan
- Department
of Neurology, National Neuroscience Institute, SGH Campus, 169856, Singapore
- Research
Department, National Neuroscience Institute, 308433, Singapore
- Neuroscience & Behavioral Disorders Program, DUKE-NUS Graduate Medical School, 169857, Singapore
| | - Li Zeng
- Neural
Stem Cell Research Lab, Research Department, National Neuroscience Institute, 308433, Singapore
- Neuroscience & Behavioral Disorders Program, DUKE-NUS Graduate Medical School, 169857, Singapore
| |
Collapse
|
174
|
Bicker S, Lackinger M, Weiß K, Schratt G. MicroRNA-132, -134, and -138: a microRNA troika rules in neuronal dendrites. Cell Mol Life Sci 2014; 71:3987-4005. [PMID: 25008044 PMCID: PMC11113804 DOI: 10.1007/s00018-014-1671-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 06/11/2014] [Accepted: 06/20/2014] [Indexed: 01/19/2023]
Abstract
Dendritic mRNA transport and local translation in the postsynaptic compartment play an important role in synaptic plasticity, learning and memory. Local protein synthesis at the synapse has to be precisely orchestrated by a plethora of factors including RNA binding proteins as well as microRNAs, an extensive class of small non-coding RNAs. By binding to complementary sequences in target mRNAs, microRNAs fine-tune protein synthesis and thereby represent critical regulators of gene expression at the post-transcriptional level. Research over the last years identified an entire network of dendritic microRNAs that fulfills an essential role in synapse development and physiology. Recent studies provide evidence that these small regulatory molecules are highly regulated themselves, at the level of expression as well as function. The importance of microRNAs for correct function of the nervous system is reflected by an increasing number of studies linking dysregulation of microRNA pathways to neurological disorders. By focusing on three extensively studied examples (miR-132, miR-134, miR-138), this review will attempt to illustrate the complex regulatory roles of dendritic microRNAs at the synapse and their implications for pathological conditions.
Collapse
Affiliation(s)
- Silvia Bicker
- Biochemical-Pharmacological Center (BPC) Marburg, Institute of Physiological Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Martin Lackinger
- Biochemical-Pharmacological Center (BPC) Marburg, Institute of Physiological Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Kerstin Weiß
- Biochemical-Pharmacological Center (BPC) Marburg, Institute of Physiological Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Gerhard Schratt
- Biochemical-Pharmacological Center (BPC) Marburg, Institute of Physiological Chemistry, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
175
|
Beneficial effects of caffeine in a transgenic model of Alzheimer's disease-like tau pathology. Neurobiol Aging 2014; 35:2079-90. [DOI: 10.1016/j.neurobiolaging.2014.03.027] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 03/09/2014] [Accepted: 03/23/2014] [Indexed: 12/27/2022]
|
176
|
MicroRNA-135a and -200b, potential Biomarkers for Alzheimer׳s disease, regulate β secretase and amyloid precursor protein. Brain Res 2014; 1583:55-64. [PMID: 25152461 DOI: 10.1016/j.brainres.2014.04.026] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 03/17/2014] [Accepted: 04/15/2014] [Indexed: 12/31/2022]
Abstract
Amyloid precursor protein (APP) and β-site amyloid precursor protein cleaving enzyme (BACE-1) play important roles in the generation of Alzheimer׳s disease (AD), a progressive neurodegenerative disorder. In the present study, microRNA (miR) microarray was used to analyze the miR expression profiles in the hippocampi from APP/PS1 transgenic and wild type mice. The miRs with significant alteration and putative targets on APP or BACE-1 were retrieved (miR-135a, -200b and -429). The deregulations of these miRs were confirmed in mice and further verified in AD patient samples by qPCR. Primary mouse hippocampal neurons, SH-SY5Y and HEK293 cells were used to study the function of miRs on APP and BACE-1. We found that miR-135a, which was downregulated significantly in hippocampi from APP/PS1 transgenic mice compared with the wild type control, directly interacted with the 3'-UTR of BACE-1 and repressed its expression and activity. On the other hand, miR-200b and -429, which were downregulated significantly in hippocampi from APP/PS1 transgenic mice compared with the wild type control, targeted the 3'-UTR of APP and repressed its expression. Furthermore, Aβ42 could downregulate miR-200b expression which may generate a vicious cycle resulted in accumulating Aβ42. The levels of miR-135a and -200b in the serum of DAT group were significantly lower than that of control groups (P<0.05). The serum miR-200b level of MCI group was higher than that of DAT group (P<0.05) and lower than that of control group (P<0.05). We also found decreased miR-135a and -200b levels in the cerebrospinal fluid of DAT group compared with the control group (P<0.05). In conclusion, these findings showed that miR-135a, -200b and -429 may take part in the progress of AD; miR-200b was of great potential as noninvasive and easily detected blood-based biomarkers of MCI and DAT patients.
Collapse
|
177
|
Luo H, Wu Q, Ye X, Xiong Y, Zhu J, Xu J, Diao Y, Zhang D, Wang M, Qiu J, Miao J, Zhang W, Wan J. Genome-wide analysis of miRNA signature in the APPswe/PS1ΔE9 mouse model of alzheimer's disease. PLoS One 2014; 9:e101725. [PMID: 25148207 PMCID: PMC4141691 DOI: 10.1371/journal.pone.0101725] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 06/10/2014] [Indexed: 12/26/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. One of the pathological hallmarks of AD is amyloid β (Aβ) deposition. MicroRNAs (miRNAs) are small non-coding RNAs whose expression levels change significantly during neuronal pathogenesis and may be used as diagnostic markers. Some miRNAs are important in AD development by targeting genes responsible for Aβ metabolism. However, a systematic assessment of the miRNA expression profile induced by Aβ-mediated neuronal pathogenesis is still lacking. In the present study, we examined miRNA expression profile by using the APPswe/PS1ΔE9 mouse model of AD. Two sibling pairs of mice were examined, showing 30 and 24 miRNAs with significantly altered expression levels from each paired control, respectively. Nine known miRNAs were common in both groups. Prediction of putative target genes and functional annotation implied that these altered miRNAs affect many target genes mainly involved in PI3K/Akt signaling pathway. This study provides a general profile of miRNAs regulated by Aβ-associated signal pathways, which is helpful to understand the mechanism of Aβ-induced neuronal dysfunction in AD development.
Collapse
Affiliation(s)
- Hongxue Luo
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Qi Wu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Xiaoyang Ye
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Yi Xiong
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Jinyong Zhu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Junyu Xu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Yarui Diao
- Ludwig Institute for Cancer Research, La Jolla, California, United States of America
| | - Duo Zhang
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Maosheng Wang
- GaoZhou people's Hospital, Gao Zhou, Guangdong Province, China
| | - Jinhua Qiu
- GaoZhou people's Hospital, Gao Zhou, Guangdong Province, China
| | - Jianting Miao
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an City, Shanxi Province, China
| | - Wei Zhang
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Jun Wan
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| |
Collapse
|
178
|
Liu CG, Song J, Zhang YQ, Wang PC. MicroRNA-193b is a regulator of amyloid precursor protein in the blood and cerebrospinal fluid derived exosomal microRNA-193b is a biomarker of Alzheimer's disease. Mol Med Rep 2014; 10:2395-400. [PMID: 25119742 DOI: 10.3892/mmr.2014.2484] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 06/24/2014] [Indexed: 11/06/2022] Open
Abstract
Amyloid precursor protein (APP) has an important function in the generation of Alzheimer's disease (AD). In our previous study, miR‑193b was found to be downregulated in the hippocampi of 9‑month‑old APP/PS1 double‑transgenic mice using microRNA (miR) array. In the present study, bioinformatic analyses showed that miR‑193b was a miR that was predicted to potentially target the 3'‑untranslated region (UTR) of APP. Subsequently, the function of miR‑193b on APP was studied. The levels of miR‑193b, exosomal miR‑193b, Aβ, tau, p‑tau, HCY and APOE in samples from APP/PS1 double‑transgenic mice, mild cognitive impairment (MCI) and dementia of Alzheimer‑type (DAT) patients, were measured. The results indicated that overexpression of miR‑193b could repress the mRNA and protein expression of APP. The miR‑193b inhibitor oligonucleotide induced upregulation of APP. Binding sites of miR‑193b in the 3'‑UTR of APP were identified by luciferase assay. MCI and DAT patients had lower exosomal miR‑193b, but not total miR‑193b, in the blood as compared with the controls. DAT patients had lower exosomal miR‑193b levels in blood as compared with the MCI group. A decreased exosomal miR‑193b expression level was additionally observed in the cerebral spinal fluid (CSF) of DAT patients. Negative correlations were found between exosomal miR‑193b and Aβ42 in the CSF of DAT patients. In conclusion, these findings showed that miR‑193b may function in the development of AD and exosomal miR‑193b has potential as a novel, non-invasive, blood‑based biomarker of MCI and DAT patients.
Collapse
Affiliation(s)
- Chen-Geng Liu
- Department of Clinical Laboratory, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Jing Song
- Department of Clinical Laboratory, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Yue-Qi Zhang
- Department of Clinical Laboratory, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Pei-Chang Wang
- Department of Clinical Laboratory, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| |
Collapse
|
179
|
Banzhaf-Strathmann J, Benito E, May S, Arzberger T, Tahirovic S, Kretzschmar H, Fischer A, Edbauer D. MicroRNA-125b induces tau hyperphosphorylation and cognitive deficits in Alzheimer's disease. EMBO J 2014; 33:1667-80. [PMID: 25001178 DOI: 10.15252/embj.201387576] [Citation(s) in RCA: 241] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Sporadic Alzheimer's disease (AD) is the most prevalent form of dementia, but no clear disease-initiating mechanism is known. Aβ deposits and neuronal tangles composed of hyperphosphorylated tau are characteristic for AD. Here, we analyze the contribution of microRNA-125b (miR-125b), which is elevated in AD. In primary neurons, overexpression of miR-125b causes tau hyperphosphorylation and an upregulation of p35, cdk5, and p44/42-MAPK signaling. In parallel, the phosphatases DUSP6 and PPP1CA and the anti-apoptotic factor Bcl-W are downregulated as direct targets of miR-125b. Knockdown of these phosphatases induces tau hyperphosphorylation, and overexpression of PPP1CA and Bcl-W prevents miR-125b-induced tau phosphorylation, suggesting that they mediate the effects of miR-125b on tau. Conversely, suppression of miR-125b in neurons by tough decoys reduces tau phosphorylation and kinase expression/activity. Injecting miR-125b into the hippocampus of mice impairs associative learning and is accompanied by downregulation of Bcl-W, DUSP6, and PPP1CA, resulting in increased tau phosphorylation in vivo. Importantly, DUSP6 and PPP1CA are also reduced in AD brains. These data implicate miR-125b in the pathogenesis of AD by promoting pathological tau phosphorylation.
Collapse
Affiliation(s)
| | - Eva Benito
- German Center for Neurodegenerative Diseases, c/o European Neuroscience Institute ENI-G, Göttingen, Germany
| | - Stephanie May
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Thomas Arzberger
- German Center for Neurodegenerative Diseases, Munich, Germany Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | - Hans Kretzschmar
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - André Fischer
- German Center for Neurodegenerative Diseases, c/o European Neuroscience Institute ENI-G, Göttingen, Germany Department of Psychiatry and Psychotherapy, University Medical Center, University Göttingen, Göttingen, Germany
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases, Munich, Germany Adolf Butenandt Institute, Biochemistry, Ludwig-Maximilians-University Munich, Munich, Germany Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
180
|
Genetic markers for diagnosis and pathogenesis of Alzheimer's disease. Gene 2014; 545:185-93. [DOI: 10.1016/j.gene.2014.05.031] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 04/07/2014] [Accepted: 05/13/2014] [Indexed: 02/06/2023]
|
181
|
Expression of Nampt in hippocampal and cortical excitatory neurons is critical for cognitive function. J Neurosci 2014; 34:5800-15. [PMID: 24760840 DOI: 10.1523/jneurosci.4730-13.2014] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD(+)) is an enzyme cofactor or cosubstrate in many essential biological pathways. To date, the primary source of neuronal NAD(+) has been unclear. NAD(+) can be synthesized from several different precursors, among which nicotinamide is the substrate predominantly used in mammals. The rate-limiting step in the NAD(+) biosynthetic pathway from nicotinamide is performed by nicotinamide phosphoribosyltransferase (Nampt). Here, we tested the hypothesis that neurons use intracellular Nampt-mediated NAD(+) biosynthesis by generating and evaluating mice lacking Nampt in forebrain excitatory neurons (CaMKIIαNampt(-/-) mice). CaMKIIαNampt(-/-) mice showed hippocampal and cortical atrophy, astrogliosis, microgliosis, and abnormal CA1 dendritic morphology by 2-3 months of age. Importantly, these histological changes occurred with altered intrahippocampal connectivity and abnormal behavior; including hyperactivity, some defects in motor skills, memory impairment, and reduced anxiety, but in the absence of impaired sensory processes or long-term potentiation of the Schaffer collateral pathway. These results clearly demonstrate that forebrain excitatory neurons mainly use intracellular Nampt-mediated NAD(+) biosynthesis to mediate their survival and function. Studying this particular NAD(+) biosynthetic pathway in these neurons provides critical insight into their vulnerability to pathophysiological stimuli and the development of therapeutic and preventive interventions for their preservation.
Collapse
|
182
|
|
183
|
Balazs R. Epigenetic mechanisms in Alzheimer's disease. Degener Neurol Neuromuscul Dis 2014; 4:85-102. [PMID: 32669903 PMCID: PMC7337154 DOI: 10.2147/dnnd.s37341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/19/2014] [Indexed: 11/25/2022] Open
Abstract
The worldwide increase in life expectancy is leading to an increase in age-dependent diseases, including nonfamilial, sporadic Alzheimer’s disease (AD), which is the subject of this review. The etiology and pathophysiology of the disease is not fully understood, but present observations suggest that, in addition to genetic risk factors, environmental influences may be involved via epigenetic mechanisms. Currently, there is no effective treatment, but there are indications that lifestyle has an impact on the development of the disease. This view is supported by preclinical studies not only showing that human lifestyle-equivalent interventions have a positive effect on cognitive function in animal models of AD, but also indicating the involvement of underlying epigenetic mechanisms. After a brief overview of the most characteristic chromatin modifications, ie, DNA methylation and histone modifications, epigenetic changes associated with aging are considered, given that aging is the most important risk factor for AD. This is followed by a description of some epigenetic alterations recognized in AD. The impact of environmental factors and lifestyle on the epigenome is then considered. Epigenetic treatments with HDAC inhibitors and RNA-based drugs are considered, which – while still in preclinical stages – are promising for potential benefit. It is concluded that while awaiting results from clinical trials in progress, focusing on lifestyle adjustments with an epigenetic background are the best way to prevent/delay the onset of this devastating disease.
Collapse
Affiliation(s)
- Robert Balazs
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| |
Collapse
|
184
|
Babski J, Maier LK, Heyer R, Jaschinski K, Prasse D, Jäger D, Randau L, Schmitz RA, Marchfelder A, Soppa J. Small regulatory RNAs in Archaea. RNA Biol 2014; 11:484-93. [PMID: 24755959 PMCID: PMC4152357 DOI: 10.4161/rna.28452] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Small regulatory RNAs (sRNAs) are universally distributed in all three domains of life, Archaea, Bacteria, and Eukaryotes. In bacteria, sRNAs typically function by binding near the translation start site of their target mRNAs and thereby inhibit or activate translation. In eukaryotes, miRNAs and siRNAs typically bind to the 3′-untranslated region (3′-UTR) of their target mRNAs and influence translation efficiency and/or mRNA stability. In archaea, sRNAs have been identified in all species investigated using bioinformatic approaches, RNomics, and RNA-Seq. Their size can vary significantly between less than 50 to more than 500 nucleotides. Differential expression of sRNA genes has been studied using northern blot analysis, microarrays, and RNA-Seq. In addition, biological functions have been unraveled by genetic approaches, i.e., by characterization of designed mutants. As in bacteria, it was revealed that archaeal sRNAs are involved in many biological processes, including metabolic regulation, adaptation to extreme conditions, stress responses, and even in regulation of morphology and cellular behavior. Recently, the first target mRNAs were identified in archaea, including one sRNA that binds to the 5′-region of two mRNAs in Methanosarcina mazei Gö1 and a few sRNAs that bind to 3′-UTRs in Sulfolobus solfataricus, three Pyrobaculum species, and Haloferax volcanii, indicating that archaeal sRNAs appear to be able to target both the 5′-UTR or the 3′-UTRs of their respective target mRNAs. In addition, archaea contain tRNA-derived fragments (tRFs), and one tRF has been identified as a major ribosome-binding sRNA in H. volcanii, which downregulates translation in response to stress. Besides regulatory sRNAs, archaea contain further classes of sRNAs, e.g., CRISPR RNAs (crRNAs) and snoRNAs.
Collapse
Affiliation(s)
- Julia Babski
- Institute for Molecular Biosciences; Biocentre; Goethe University; Frankfurt, Germany
| | | | - Ruth Heyer
- Biology II; Ulm University; Ulm, Germany
| | - Katharina Jaschinski
- Institute for Molecular Biosciences; Biocentre; Goethe University; Frankfurt, Germany
| | - Daniela Prasse
- Institute for Microbiology; Christian-Albrechts-University; Kiel, Germany
| | - Dominik Jäger
- Institute for Microbiology; Christian-Albrechts-University; Kiel, Germany
| | - Lennart Randau
- Prokaryotic Small RNA Biology Group; Max Planck Institute for Terrestrial Microbiology; Marburg, Germany
| | - Ruth A Schmitz
- Institute for Microbiology; Christian-Albrechts-University; Kiel, Germany
| | | | - Jörg Soppa
- Institute for Molecular Biosciences; Biocentre; Goethe University; Frankfurt, Germany
| |
Collapse
|
185
|
Generation and phenotyping of a collection of sRNA gene deletion mutants of the haloarchaeon Haloferax volcanii. PLoS One 2014; 9:e90763. [PMID: 24637842 PMCID: PMC3956466 DOI: 10.1371/journal.pone.0090763] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 02/04/2014] [Indexed: 11/19/2022] Open
Abstract
The haloarchaeon Haloferax volcanii was shown to contain 145 intergenic and 45 antisense sRNAs. In a comprehensive approach to unravel various biological roles of haloarchaeal sRNAs in vivo, 27 sRNA genes were selected and deletion mutants were generated. The phenotypes of these mutants were compared to that of the parent strain under ten different conditions, i.e. growth on four different carbon sources, growth at three different salt concentrations, and application of four different stress conditions. In addition, cell morphologies in exponential and stationary phase were observed. Furthermore, swarming of 17 mutants was analyzed. 24 of the 27 mutants exhibited a difference from the parent strain under at least one condition, revealing that haloarchaeal sRNAs are involved in metabolic regulation, growth under extreme conditions, regulation of morphology and behavior, and stress adaptation. Notably, 7 deletion mutants showed a gain of function phenotype, which has not yet been described for any other prokaryotic sRNA gene deletion mutant. Comparison of the transcriptomes of one sRNA gene deletion mutant and the parent strain led to the identification of differentially expressed genes. Genes for flagellins and chemotaxis were up-regulated in the mutant, in accordance with its gain of function swarming phenotype. While the deletion mutant analysis underscored that haloarchaeal sRNAs are involved in many biological functions, the degree of conservation is extremely low. Only 3 of the 27 genes are conserved in more than 10 haloarchaeal species. 22 of the 27 genes are confined to H. volcanii, indicating a fast evolution of haloarchaeal sRNA genes.
Collapse
|
186
|
Barbato C, Pezzola S, Caggiano C, Antonelli M, Frisone P, Ciotti MT, Ruberti F. A lentiviral sponge for miR-101 regulates RanBP9 expression and amyloid precursor protein metabolism in hippocampal neurons. Front Cell Neurosci 2014; 8:37. [PMID: 24592211 PMCID: PMC3923151 DOI: 10.3389/fncel.2014.00037] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 01/25/2014] [Indexed: 12/22/2022] Open
Abstract
Neurodegeneration associated with amyloid β (Aβ) peptide accumulation, synaptic loss, and memory impairment are pathophysiological features of Alzheimer's disease (AD). Numerous microRNAs regulate amyloid precursor protein (APP) expression and metabolism. We previously reported that miR-101 is a negative regulator of APP expression in cultured hippocampal neurons. In this study, a search for predicted APP metabolism-associated miR-101 targets led to the identification of a conserved miR-101 binding site within the 3′ untranslated region (UTR) of the mRNA encoding Ran-binding protein 9 (RanBP9). RanBP9 increases APP processing by β-amyloid converting enzyme 1 (BACE1), secretion of soluble APPβ (sAPPβ), and generation of Aβ. MiR-101 significantly reduced reporter gene expression when co-transfected with a RanBP9 3′-UTR reporter construct, while site-directed mutagenesis of the predicted miR-101 target site eliminated the reporter response. To investigate the effect of stable inhibition of miR-101 both in vitro and in vivo, a microRNA sponge was developed to bind miR-101 and derepress its targets. Four tandem bulged miR-101 responsive elements (REs), located downstream of the enhanced green fluorescence protein (EGFP) open reading frame and driven by the synapsin promoter, were placed in a lentiviral vector to create the pLSyn-miR-101 sponge. Delivery of the sponge to primary hippocampal neurons significantly increased both APP and RanBP9 expression, as well as sAPPβ levels in the conditioned medium. Importantly, silencing of endogenous RanBP9 reduced sAPPβ levels in miR-101 sponge-containing hippocampal cultures, indicating that miR-101 inhibition may increase amyloidogenic processing of APP by RanBP9. Lastly, the impact of miR-101 on its targets was demonstrated in vivo by intrahippocampal injection of the pLSyn-miR-101 sponge into C57BL6 mice. This study thus provides the basis for studying the consequences of long-term miR-101 inhibition on the pathology of AD.
Collapse
Affiliation(s)
- Christian Barbato
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR) Rome, Italy
| | - Silvia Pezzola
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR) Rome, Italy
| | - Cinzia Caggiano
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR) Rome, Italy
| | - Martina Antonelli
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR) Rome, Italy
| | - Paola Frisone
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR) Rome, Italy
| | - Maria Teresa Ciotti
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR) Rome, Italy
| | - Francesca Ruberti
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR) Rome, Italy
| |
Collapse
|
187
|
Barca-Mayo O, De Pietri Tonelli D. Convergent microRNA actions coordinate neocortical development. Cell Mol Life Sci 2014; 71:2975-95. [PMID: 24519472 PMCID: PMC4111863 DOI: 10.1007/s00018-014-1576-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 01/11/2014] [Accepted: 01/27/2014] [Indexed: 12/19/2022]
Abstract
Neocortical development is a complex process that, at the cellular level, involves tight control of self-renewal, cell fate commitment, survival, differentiation and delamination/migration. These processes require, at the molecular level, the precise regulation of intrinsic signaling pathways and extrinsic factors with coordinated action in a spatially and temporally specific manner. Transcriptional regulation plays an important role during corticogenesis; however, microRNAs (miRNAs) are emerging as important post-transcriptional regulators of various aspects of central nervous system development. miRNAs are a class of small, single-stranded noncoding RNA molecules that control the expression of the majority of protein coding genes (i.e., targets). How do different miRNAs achieve precise control of gene networks during neocortical development? Here, we critically review all the miRNA–target interactions validated in vivo, with relevance to the generation and migration of pyramidal-projection glutamatergic neurons, and for the initial formation of cortical layers in the embryonic development of rodent neocortex. In particular, we focus on convergent miRNA actions, which are still a poorly understood layer of complexity in miRNA signaling, but potentially one of the keys to disclosing how miRNAs achieve the precise coordination of complex biological processes such as neocortical development.
Collapse
Affiliation(s)
- Olga Barca-Mayo
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genoa, Italy
| | | |
Collapse
|
188
|
Gene and MicroRNA transcriptome analysis of Parkinson's related LRRK2 mouse models. PLoS One 2014; 9:e85510. [PMID: 24427314 PMCID: PMC3888428 DOI: 10.1371/journal.pone.0085510] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 11/28/2013] [Indexed: 12/12/2022] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most frequent cause of genetic Parkinson’s disease (PD). The biological function of LRRK2 and how mutations lead to disease remain poorly defined. It has been proposed that LRRK2 could function in gene transcription regulation; however, this issue remains controversial. Here, we investigated in parallel gene and microRNA (miRNA) transcriptome profiles of three different LRRK2 mouse models. Striatal tissue was isolated from adult LRRK2 knockout (KO) mice, as well as mice expressing human LRRK2 wildtype (hLRRK2-WT) or the PD-associated R1441G mutation (hLRRK2-R1441G). We identified a total of 761 genes and 24 miRNAs that were misregulated in the absence of LRRK2 when a false discovery rate of 0.2 was applied. Notably, most changes in gene expression were modest (i.e., <2 fold). By real-time quantitative RT-PCR, we confirmed the variations of selected genes (e.g., adra2, syt2, opalin) and miRNAs (e.g., miR-16, miR-25). Surprisingly, little or no changes in gene expression were observed in mice expressing hLRRK2-WT or hLRRK2-R1441G when compared to non-transgenic controls. Nevertheless, a number of miRNAs were misexpressed in these models. Bioinformatics analysis identified several miRNA-dependent and independent networks dysregulated in LRRK2-deficient mice, including PD-related pathways. These results suggest that brain LRRK2 plays an overall modest role in gene transcription regulation in mammals; however, these effects seem context and RNA type-dependent. Our data thus set the stage for future investigations regarding LRRK2 function in PD development.
Collapse
|
189
|
Restricted perinatal retinal degeneration induces retina reshaping and correlated structural rearrangement of the retinotopic map. Nat Commun 2013; 4:1938. [PMID: 23733098 PMCID: PMC3709497 DOI: 10.1038/ncomms2926] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 04/24/2013] [Indexed: 02/05/2023] Open
Abstract
The formation of the retinotopic map depends on the action of axon guidance molecules, activity-dependent mechanisms and axonal competition. However, little is known about the plasticity potential of the system and the effects on the remodelling of retinocollicular connections upon retinal insults. Here we create a mouse model in which retinal ganglion cells that project to anterior and posterior superior colliculus undergo cell death during topographic map formation. We show that the remaining retinal ganglion cells expand the targeted area in the superior colliculus and at the same time increase their spatial coverage in the retina in a correlated fashion. The resulting contralateral topographic map is overall maintained but less precise, while ipsilateral retinal ganglion cell axons are abnormally distributed in anterior and posterior superficial superior colliculus. These results suggest the presence of plastic mechanisms in the developing mammalian visual system to adjust retinal space and its target coverage and ensure a uniform map.
Collapse
|
190
|
Meng F, Dai E, Yu X, Zhang Y, Chen X, Liu X, Wang S, Wang L, Jiang W. Constructing and characterizing a bioactive small molecule and microRNA association network for Alzheimer's disease. J R Soc Interface 2013; 11:20131057. [PMID: 24352679 DOI: 10.1098/rsif.2013.1057] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disorder. Much effort has been devoted to developing effective therapeutic agents. Recently, targeting microRNAs (miRNAs) with small molecules has become a novel therapy for human diseases. In this study, we present a systematic computational approach to construct a bioactive Small molecule and miRNA association Network in AD (SmiRN-AD), which is based on the gene expression signatures of bioactive small molecule perturbation and AD-related miRNA regulation. We also performed topological and functional analysis of the SmiRN-AD from multiple perspectives. At the significance level of p ≤ 0.01, 496 small molecule-miRNA associations, including 25 AD-related miRNAs and 275 small molecules, were recognized and used to construct the SmiRN-AD. The drugs that were connected with the same miRNA tended to share common drug targets (p = 1.72 × 10(-4)) and belong to the same therapeutic category (p = 4.22 × 10(-8)). The miRNAs that were linked to the same small molecule regulated more common miRNA targets (p = 6.07 × 10(-3)). Further analysis of the positive connections (quinostatin and miR-148b, amantadine and miR-15a) and the negative connections (melatonin and miR-30e-5p) indicated that our large-scale predictions afforded specific biological insights into AD pathogenesis and therapy. This study proposes a holistic strategy for deciphering the associations between small molecules and miRNAs in AD, which may be helpful for developing a novel effective miRNA-associated therapeutic strategy for AD. A comprehensive database for the SmiRN-AD and the differential expression patterns of the miRNA targets in AD is freely available at http://bioinfo.hrbmu.edu.cn/SmiRN-AD/.
Collapse
Affiliation(s)
- Fanlin Meng
- College of Bioinformatics Science and Technology, Harbin Medical University, , 194 Xuefu Road, Harbin 150081, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Heman-Ackah SM, Hallegger M, Rao MS, Wood MJA. RISC in PD: the impact of microRNAs in Parkinson's disease cellular and molecular pathogenesis. Front Mol Neurosci 2013; 6:40. [PMID: 24312000 PMCID: PMC3834244 DOI: 10.3389/fnmol.2013.00040] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/31/2013] [Indexed: 12/19/2022] Open
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative disease characterized primarily by the selective death of dopaminergic (DA) neurons in the substantia nigra pars compacta of the midbrain. Although several genetic forms of PD have been identified, the precise molecular mechanisms underlying DA neuron loss in PD remain elusive. In recent years, microRNAs (miRNAs) have been recognized as potent post-transcriptional regulators of gene expression with fundamental roles in numerous biological processes. Although their role in PD pathogenesis is still a very active area of investigation, several seminal studies have contributed significantly to our understanding of the roles these small non-coding RNAs play in the disease process. Among these are studies which have demonstrated specific miRNAs that target and down-regulate the expression of PD-related genes as well as those demonstrating a reciprocal relationship in which PD-related genes act to regulate miRNA processing machinery. Concurrently, a wealth of knowledge has become available regarding the molecular mechanisms that unify the underlying etiology of genetic and sporadic PD pathogenesis, including dysregulated protein quality control by the ubiquitin-proteasome system and autophagy pathway, activation of programmed cell death, mitochondrial damage and aberrant DA neurodevelopment and maintenance. Following a discussion of the interactions between PD-related genes and miRNAs, this review highlights those studies which have elucidated the roles of these pathways in PD pathogenesis. We highlight the potential of miRNAs to serve a critical regulatory role in the implicated disease pathways, given their capacity to modulate the expression of entire families of related genes. Although few studies have directly linked miRNA regulation of these pathways to PD, a strong foundation for investigation has been laid and this area holds promise to reveal novel therapeutic targets for PD.
Collapse
Affiliation(s)
- Sabrina M Heman-Ackah
- Department of Physiology, Anatomy and Genetics, University of Oxford Oxford, UK ; Center for Regenerative Medicine, US National Institutes of Health Bethesda, MD, USA
| | | | | | | |
Collapse
|
192
|
Dicer expression is essential for adult midbrain dopaminergic neuron maintenance and survival. Mol Cell Neurosci 2013; 58:22-8. [PMID: 24184162 DOI: 10.1016/j.mcn.2013.10.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 08/20/2013] [Accepted: 10/23/2013] [Indexed: 01/28/2023] Open
Abstract
The type III RNAse, Dicer, is responsible for the processing of microRNA (miRNA) precursors into functional miRNA molecules, non-coding RNAs that bind to and target messenger RNAs for repression. Dicer expression is essential for mouse midbrain development and dopaminergic (DAergic) neuron maintenance and survival during the early post-natal period. However, the role of Dicer in adult mouse DAergic neuron maintenance and survival is unknown. To bridge this gap in knowledge, we selectively knocked-down Dicer expression in individual DAergic midbrain areas, including the ventral tegmental area (VTA) and substantia nigra pars compacta (SNpc) via viral-mediated expression of Cre in adult floxed Dicer knock-in mice (Dicer(flox/flox)). Bilateral Dicer loss in the VTA resulted in progressive hyperactivity that was significantly reduced by the dopamine agonist, amphetamine. In contrast, decreased Dicer expression in the SNpc did not affect locomotor activity but did induce motor-learning impairment on an accelerating rotarod. Knock-down of Dicer in both midbrain regions of adult Dicer(flox/flox) mice resulted in preferential, progressive loss of DAergic neurons likely explaining motor behavior phenotypes. In addition, knock-down of Dicer in midbrain areas triggered neuronal death via apoptosis. Together, these data indicate that Dicer expression and, as a consequence, miRNA function, are essential for DAergic neuronal maintenance and survival in adult midbrain DAergic neuron brain areas.
Collapse
|
193
|
Label-free voltammetric detection of MicroRNAs at multi-channel screen printed array of electrodes comparison to graphite sensors. Talanta 2013; 118:7-13. [PMID: 24274264 DOI: 10.1016/j.talanta.2013.09.041] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 09/15/2013] [Accepted: 09/20/2013] [Indexed: 01/19/2023]
Abstract
The multi-channel screen-printed array of electrodes (MUX-SPE16) was used in our study for the first time for electrochemical monitoring of nucleic acid hybridization related to different miRNA sequences (miRNA-16, miRNA-15a and miRNA-660, i.e, the biomarkers for Alzheimer disease). The MUX-SPE16 was also used for the first time herein for the label-free electrochemical detection of nucleic acid hybridization combined magnetic beads (MB) assay in comparison to the disposable pencil graphite electrode (PGE). Under the principle of the magnetic beads assay, the biotinylated inosine substituted DNA probe was firstly immobilized onto streptavidin coated MB, and then, the hybridization process between probe and its complementary miRNA sequence was performed at MB surface. The voltammetric transduction was performed using differential pulse voltammetry (DPV) technique in combination with the single-use graphite sensor technologies; PGE and MUX-SPE16 for miRNA detection by measuring the guanine oxidation signal without using any external indicator. The features of single-use sensor technologies, PGE and MUX-SPE16, were discussed concerning to their reproducibility, detection limit, and selectivity compared to the results in the earlier studies presenting the electrochemical miRNA detection related to different miRNA sequences.
Collapse
|
194
|
Veerappan CS, Sleiman S, Coppola G. Epigenetics of Alzheimer's disease and frontotemporal dementia. Neurotherapeutics 2013; 10:709-21. [PMID: 24150812 PMCID: PMC3805876 DOI: 10.1007/s13311-013-0219-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This article will review the recent advances in the understanding of the role of epigenetic modifications and the promise of future epigenetic therapy in neurodegenerative dementias, including Alzheimer's disease and frontotemporal dementia.
Collapse
Affiliation(s)
- Chendhore S Veerappan
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA,
| | | | | |
Collapse
|
195
|
Hong J, Zhang H, Kawase-Koga Y, Sun T. MicroRNA function is required for neurite outgrowth of mature neurons in the mouse postnatal cerebral cortex. Front Cell Neurosci 2013; 7:151. [PMID: 24062642 PMCID: PMC3772315 DOI: 10.3389/fncel.2013.00151] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/25/2013] [Indexed: 12/18/2022] Open
Abstract
The structure of the postnatal mammalian cerebral cortex is an assembly of numerous mature neurons that exhibit proper neurite outgrowth and axonal and dendritic morphology. While many protein coding genes are shown to be involved in neuronal maturation, the role of microRNAs (miRNAs) in this process is also becoming evident. We here report that blocking miRNA biogenesis in differentiated neurons results in microcephaly like phenotypes in the postnatal mouse brain. The smaller brain defect is not caused by defective neurogenesis, altered neuronal migration or significant neuronal cell death. Surprisingly, a dramatic increase in neuronal packing density within the postnatal brain is observed. Loss of miRNA function causes shorter neurite outgrowth and smaller soma size of mature neurons in vitro. Our results reveal the impact of miRNAs on normal development of neuronal morphology and brain function. Because neurite outgrowth is critical for neuroregeneration, our studies further highlight the importance of miRNAs in the treatment of neurological diseases.
Collapse
Affiliation(s)
- Janet Hong
- Department of Cell and Developmental Biology, Cornell University Weill Medical College New York, NY, USA
| | | | | | | |
Collapse
|
196
|
Wang J, Yu JT, Tan MS, Jiang T, Tan L. Epigenetic mechanisms in Alzheimer's disease: implications for pathogenesis and therapy. Ageing Res Rev 2013; 12:1024-41. [PMID: 23688931 DOI: 10.1016/j.arr.2013.05.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 05/08/2013] [Indexed: 12/14/2022]
Abstract
The vast majority of Alzheimer's disease (AD) are late-onset forms (LOAD) likely due to the interplay of environmental influences and individual genetic susceptibility. Epigenetic mechanisms, including DNA methylation, histone modifications and non-coding RNAs, constitute dynamic intracellular processes for translating environmental stimuli into modifications in gene expression. Over the past decade it has become increasingly clear that epigenetic mechanisms play a pivotal role in aging the pathogenesis of AD. Here, we provide a review of the major mechanisms for epigenetic modification and how they are reportedly altered in aging and AD. Moreover, we also consider how aberrant epigenetic modifications may lead to AD pathogenesis, and we review the therapeutic potential of epigenetic treatments for AD.
Collapse
Affiliation(s)
- Jun Wang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, China
| | | | | | | | | |
Collapse
|
197
|
Dorval V, Nelson PT, Hébert SS. Circulating microRNAs in Alzheimer's disease: the search for novel biomarkers. Front Mol Neurosci 2013; 6:24. [PMID: 24009553 PMCID: PMC3757537 DOI: 10.3389/fnmol.2013.00024] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 08/11/2013] [Indexed: 01/28/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disease in the elderly. While advancements have been made in understanding the genetic and molecular basis of AD, the clinical diagnosis of AD remains difficult, and post-mortem confirmation is often required. Furthermore, the onset of neurodegeneration precedes clinical symptoms by approximately a decade. Consequently, there is a crucial need for an early and accurate diagnosis of AD, which can potentially lead to strategies that can slow down or stop the progression of neurodegeneration and dementia. Recent advances in the non-coding RNA field have shown that microRNAs (miRNAs) can function as powerful biomarkers in human diseases. Studies are emerging suggesting that circulating miRNAs in the cerebrospinal fluid and blood serum have characteristic changes in AD patients. Whether miRNAs can be used in AD diagnosis, alone or in combination with other AD biomarkers (e.g., amyloid and tau), warrants further investigation.
Collapse
Affiliation(s)
- Véronique Dorval
- Axe Neurosciences, Centre de Recherche du Centre Hospitalier Universitaire de Québec (Centre Hospitalier de l'Université Laval), Québec QC, Canada ; Département de Psychiatrie et de Neurosciences, Université Laval, Québec QC, Canada
| | | | | |
Collapse
|
198
|
Abstract
Epigenetics describes heritable alterations of gene expression that do not involve DNA sequence variation and are changeable throughout an organism's lifetime. Not only can epigenetic status influence drug response, but it can also be modulated by drugs. In this review, the three major epigenetic mechanisms are described: covalent DNA modification, histone protein modification, and regulation by noncoding RNA. Further, this review describes how drug therapy can influence, and be influenced by, these mechanisms. Drugs with epigenetic mechanisms are already in use, with many more likely to be approved within the next few years. As the understanding of epigenetic processes improves, so will the ability to use these data in the clinic to improve patient care.
Collapse
Affiliation(s)
- Julio D Duarte
- Department of Pharmacy Practice, Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
199
|
Fragile X-associated tremor/ataxia syndrome (FXTAS): pathology and mechanisms. Acta Neuropathol 2013; 126:1-19. [PMID: 23793382 DOI: 10.1007/s00401-013-1138-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 05/30/2013] [Indexed: 12/17/2022]
Abstract
Since its discovery in 2001, our understanding of fragile X-associated tremor/ataxia syndrome (FXTAS) has undergone a remarkable transformation. Initially characterized rather narrowly as an adult-onset movement disorder, the definition of FXTAS is broadening; moreover, the disorder is now recognized as only one facet of a much broader clinical pleiotropy among children and adults who carry premutation alleles of the FMR1 gene. Furthermore, the intranuclear inclusions of FXTAS, once thought to be a CNS-specific marker of the disorder, are now known to be widely distributed in multiple non-CNS tissues; this observation fundamentally changes our concept of the disease, and may provide the basis for understanding the diverse medical problems associated with the premutation. Recent work on the pathogenic mechanisms underlying FXTAS indicates that the origins of the late-onset neurodegenerative disorder actually lie in early development, raising the likelihood that all forms of clinical involvement among premutation carriers have a common underlying mechanistic basis. There has also been great progress in our understanding of the triggering event(s) in FXTAS pathogenesis, which is now thought to involve sequestration of one or more nuclear proteins involved with microRNA biogenesis. Moreover, there is mounting evidence that mitochondrial dysregulation contributes to the decreased cell function and loss of viability, evident in mice even during the neonatal period. Taken together, these recent findings offer hope for early interventions for FXTAS, well before the onset of overt disease, and for the treatment of other forms of clinical involvement among premutation carriers.
Collapse
|
200
|
Sellier C, Freyermuth F, Tabet R, Tran T, He F, Ruffenach F, Alunni V, Moine H, Thibault C, Page A, Tassone F, Willemsen R, Disney MD, Hagerman PJ, Todd PK, Charlet-Berguerand N. Sequestration of DROSHA and DGCR8 by expanded CGG RNA repeats alters microRNA processing in fragile X-associated tremor/ataxia syndrome. Cell Rep 2013; 3:869-80. [PMID: 23478018 DOI: 10.1016/j.celrep.2013.02.004] [Citation(s) in RCA: 190] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 11/30/2012] [Accepted: 02/01/2013] [Indexed: 11/19/2022] Open
Abstract
Fragile X-associated tremor/ataxia syndrome (FXTAS) is an inherited neurodegenerative disorder caused by the expansion of 55-200 CGG repeats in the 5' UTR of FMR1. These expanded CGG repeats are transcribed and accumulate in nuclear RNA aggregates that sequester one or more RNA-binding proteins, thus impairing their functions. Here, we have identified that the double-stranded RNA-binding protein DGCR8 binds to expanded CGG repeats, resulting in the partial sequestration of DGCR8 and its partner, DROSHA, within CGG RNA aggregates. Consequently, the processing of microRNAs (miRNAs) is reduced, resulting in decreased levels of mature miRNAs in neuronal cells expressing expanded CGG repeats and in brain tissue from patients with FXTAS. Finally, overexpression of DGCR8 rescues the neuronal cell death induced by expression of expanded CGG repeats. These results support a model in which a human neurodegenerative disease originates from the alteration, in trans, of the miRNA-processing machinery.
Collapse
Affiliation(s)
- Chantal Sellier
- Department of Translational Medicine, IGBMC, Illkirch 67400, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|