51
|
Rahman MR, Islam T, Huq F, Quinn JM, Moni MA. Identification of molecular signatures and pathways common to blood cells and brain tissue of amyotrophic lateral sclerosis patients. INFORMATICS IN MEDICINE UNLOCKED 2019. [DOI: 10.1016/j.imu.2019.100193] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
52
|
Klein C, Roussel G, Brun S, Rusu C, Patte-Mensah C, Maitre M, Mensah-Nyagan AG. 5-HIAA induces neprilysin to ameliorate pathophysiology and symptoms in a mouse model for Alzheimer's disease. Acta Neuropathol Commun 2018; 6:136. [PMID: 30537985 PMCID: PMC6290545 DOI: 10.1186/s40478-018-0640-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 11/28/2018] [Indexed: 01/07/2023] Open
Abstract
Serotoninergic activation which decreases brain Aβ peptides is considered beneficial in mouse models for Alzheimer's disease (AD), but the mechanisms involved remain unclear. Because growing evidence suggested that the stimulation of proteases digesting Aβ, especially the endopeptidase neprilysin (NEP) may be effective for AD therapy/prevention, we explored the involvement of serotonin precursors and derivatives in NEP regulation. We found that 5-hydroxyindolacetic acid (5-HIAA), the final metabolite of serotonin, considered until now as a dead-end and inactive product of serotonin catabolism, significantly reduces brain Aβ in the transgenic APPSWE mouse model for AD-related Aβ pathology and in the phosphoramidon-induced cerebral NEP inhibition mouse model. 5-HIAA treatment improves memory performance in APPSWE mice. Furthermore, 5-HIAA and its precursors increase NEP level in vivo and in neuroblastoma cells. Inhibition of ERK 1/2 cascade by 5-HIAA or SCH772984 enhanced NEP levels, suggesting MAP-kinase pathway involvement in 5-HIAA-induced regulation of NEP expression. Our results provide the first demonstration that 5-HIAA is an active serotonin metabolite that increases brain Aβ degradation/clearance and improves symptoms in the APPSWE mouse model for AD.
Collapse
|
53
|
Benhelli-Mokrani H, Mansuroglu Z, Chauderlier A, Albaud B, Gentien D, Sommer S, Schirmer C, Laqueuvre L, Josse T, Buée L, Lefebvre B, Galas MC, Souès S, Bonnefoy E. Genome-wide identification of genic and intergenic neuronal DNA regions bound by Tau protein under physiological and stress conditions. Nucleic Acids Res 2018; 46:11405-11422. [PMID: 30321409 PMCID: PMC6265482 DOI: 10.1093/nar/gky929] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/20/2018] [Accepted: 10/04/2018] [Indexed: 01/06/2023] Open
Abstract
Tauopathies such as Alzheimer's Disease (AD) are neurodegenerative disorders for which there is presently no cure. They are named after the abnormal oligomerization/aggregation of the neuronal microtubule-associated Tau protein. Besides its role as a microtubule-associated protein, a DNA-binding capacity and a nuclear localization for Tau protein has been described in neurons. While questioning the potential role of Tau-DNA binding in the development of tauopathies, we have carried out a large-scale analysis of the interaction of Tau protein with the neuronal genome under physiological and heat stress conditions using the ChIP-on-chip technique that combines Chromatin ImmunoPrecipitation (ChIP) with DNA microarray (chip). Our findings show that Tau protein specifically interacts with genic and intergenic DNA sequences of primary culture of neurons with a preference for DNA regions positioned beyond the ±5000 bp range from transcription start site. An AG-rich DNA motif was found recurrently present within Tau-interacting regions and 30% of Tau-interacting regions overlapped DNA sequences coding for lncRNAs. Neurological processes affected in AD were enriched among Tau-interacting regions with in vivo gene expression assays being indicative of a transcriptional repressor role for Tau protein, which was exacerbated in neurons displaying nuclear pathological oligomerized forms of Tau protein.
Collapse
Affiliation(s)
- Houda Benhelli-Mokrani
- Université Paris Descartes, Centre Interdisciplinaire Chimie Biologie-Paris, Inserm UMRS1007, Paris, France
| | - Zeyni Mansuroglu
- Université Paris Descartes, Centre Interdisciplinaire Chimie Biologie-Paris, Inserm UMRS1007, Paris, France
| | - Alban Chauderlier
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172, LabEx DISTALZ, Alzheimer & Tauopathies, Lille, France
| | - Benoit Albaud
- Institut Curie, PSL Research University, Translational Research Departement, Genomics Platform, Paris, F-75248 France
| | - David Gentien
- Institut Curie, PSL Research University, Translational Research Departement, Genomics Platform, Paris, F-75248 France
| | - Sabrina Sommer
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172, LabEx DISTALZ, Alzheimer & Tauopathies, Lille, France
| | - Claire Schirmer
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172, LabEx DISTALZ, Alzheimer & Tauopathies, Lille, France
| | - Lucie Laqueuvre
- Université Paris Descartes, Centre Interdisciplinaire Chimie Biologie-Paris, Inserm UMRS1007, Paris, France
| | - Thibaut Josse
- Université François Rabelais, Institut de Recherche sur la Biologie de l’Insecte, CNRS UMR 7261, Tours, France
| | - Luc Buée
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172, LabEx DISTALZ, Alzheimer & Tauopathies, Lille, France
| | - Bruno Lefebvre
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172, LabEx DISTALZ, Alzheimer & Tauopathies, Lille, France
| | - Marie-Christine Galas
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172, LabEx DISTALZ, Alzheimer & Tauopathies, Lille, France
| | - Sylvie Souès
- Université Paris Descartes, Centre Interdisciplinaire Chimie Biologie-Paris, Inserm UMRS1007, Paris, France
| | - Eliette Bonnefoy
- Université Paris Descartes, Centre Interdisciplinaire Chimie Biologie-Paris, Inserm UMRS1007, Paris, France
| |
Collapse
|
54
|
Kramarz B, Roncaglia P, Meldal BHM, Huntley RP, Martin MJ, Orchard S, Parkinson H, Brough D, Bandopadhyay R, Hooper NM, Lovering RC. Improving the Gene Ontology Resource to Facilitate More Informative Analysis and Interpretation of Alzheimer's Disease Data. Genes (Basel) 2018; 9:E593. [PMID: 30501127 PMCID: PMC6315915 DOI: 10.3390/genes9120593] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 12/28/2022] Open
Abstract
The analysis and interpretation of high-throughput datasets relies on access to high-quality bioinformatics resources, as well as processing pipelines and analysis tools. Gene Ontology (GO, geneontology.org) is a major resource for gene enrichment analysis. The aim of this project, funded by the Alzheimer's Research United Kingdom (ARUK) foundation and led by the University College London (UCL) biocuration team, was to enhance the GO resource by developing new neurological GO terms, and use GO terms to annotate gene products associated with dementia. Specifically, proteins and protein complexes relevant to processes involving amyloid-beta and tau have been annotated and the resulting annotations are denoted in GO databases as 'ARUK-UCL'. Biological knowledge presented in the scientific literature was captured through the association of GO terms with dementia-relevant protein records; GO itself was revised, and new GO terms were added. This literature biocuration increased the number of Alzheimer's-relevant gene products that were being associated with neurological GO terms, such as 'amyloid-beta clearance' or 'learning or memory', as well as neuronal structures and their compartments. Of the total 2055 annotations that we contributed for the prioritised gene products, 526 have associated proteins and complexes with neurological GO terms. To ensure that these descriptive annotations could be provided for Alzheimer's-relevant gene products, over 70 new GO terms were created. Here, we describe how the improvements in ontology development and biocuration resulting from this initiative can benefit the scientific community and enhance the interpretation of dementia data.
Collapse
Affiliation(s)
- Barbara Kramarz
- UCL Institute of Cardiovascular Science, University College London, Rayne Building, 5 University Street, London WC1E 6JF, UK.
| | - Paola Roncaglia
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK.
| | - Birgit H M Meldal
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK.
| | - Rachael P Huntley
- UCL Institute of Cardiovascular Science, University College London, Rayne Building, 5 University Street, London WC1E 6JF, UK.
| | - Maria J Martin
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK.
| | - Sandra Orchard
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK.
| | - Helen Parkinson
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK.
| | - David Brough
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, UK.
| | - Rina Bandopadhyay
- UCL Queen Square Institute of Neurology and Reta Lila Weston Institute of Neurological Studies, 1 Wakefield Street, London WC1N 1PJ, UK.
| | - Nigel M Hooper
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, UK.
| | - Ruth C Lovering
- UCL Institute of Cardiovascular Science, University College London, Rayne Building, 5 University Street, London WC1E 6JF, UK.
| |
Collapse
|
55
|
Alibhai JD, Diack AB, Manson JC. Unravelling the glial response in the pathogenesis of Alzheimer's disease. FASEB J 2018; 32:5766-5777. [PMID: 30376380 DOI: 10.1096/fj.201801360r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease is a progressive, incurable neurodegenerative disease targeting specific neuronal populations within the brain while neighboring neurons appear unaffected. The focus for defining mechanisms has therefore been on the pathogenesis in affected neuronal populations and developing intervention strategies to prevent their cell death. However, there is growing recognition of the importance of glial cells in the development of pathology. Determining exactly how glial cells are involved in the disease process and the susceptibility of the aging brain provides unprecedented challenges. The present review examines recent studies attempting to unravel the glial response during the course of disease and how this action may dictate the outcome of neurodegeneration. The importance of regional heterogeneity of glial cells within the CNS during healthy aging and disease is examined to understand how the glial cells may contribute to neuronal susceptibility or resilience during the neurodegenerative process.-Alibhai, J. D., Diack, A. B., Manson, J. C. Unravelling the glial response in the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- James D Alibhai
- National Creutzfeldt-Jakob Disease (CJD) Research and Surveillance Unit, University of Edinburgh, Edinburgh, United Kingdom.,UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom; and
| | - Abigail B Diack
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Roslin, United Kingdom
| | - Jean C Manson
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
56
|
Taalab YM, Ibrahim N, Maher A, Hassan M, Mohamed W, Moustafa AA, Salama M, Johar D, Bernstein L. Mechanisms of disordered neurodegenerative function: concepts and facts about the different roles of the protein kinase RNA-like endoplasmic reticulum kinase (PERK). Rev Neurosci 2018; 29:387-415. [PMID: 29303785 DOI: 10.1515/revneuro-2017-0071] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/20/2017] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases, such as Alzheimer's disease, Huntington's disease, Parkinson's disease, prion disease, and amyotrophic lateral sclerosis, are a dissimilar group of disorders that share a hallmark feature of accumulation of abnormal intraneuronal or extraneuronal misfolded/unfolded protein and are classified as protein misfolding disorders. Cellular and endoplasmic reticulum (ER) stress activates multiple signaling cascades of the unfolded protein response (UPR). Consequently, translational and transcriptional alterations in target gene expression occur in response directed toward restoring the ER capacity of proteostasis and reestablishing the cellular homeostasis. Evidences from in vitro and in vivo disease models indicate that disruption of ER homeostasis causes abnormal protein aggregation that leads to synaptic and neuronal dysfunction. However, the exact mechanism by which it contributes to disease progression and pathophysiological changes remains vague. Downstream signaling pathways of UPR are fully integrated, yet with diverse unexpected outcomes in different disease models. Three well-identified ER stress sensors have been implicated in UPR, namely, inositol requiring enzyme 1, protein kinase RNA-activated-like ER kinase (PERK), and activating transcription factor 6. Although it cannot be denied that each of the involved stress sensor initiates a distinct downstream signaling pathway, it becomes increasingly clear that shared pathways are crucial in determining whether or not the UPR will guide the cells toward adaptive prosurvival or proapoptotic responses. We review a body of work on the mechanism of neurodegenerative diseases based on oxidative stress and cell death pathways with emphasis on the role of PERK.
Collapse
Affiliation(s)
- Yasmeen M Taalab
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Al-Mansoura University, Al-Mansoura, 35111, Egypt
| | - Nour Ibrahim
- Faculty of Medicine, Ain Shams University, Cairo, 11591, Egypt
| | - Ahmed Maher
- Zoonotic Disease Department, National Research Center, Dokki, Giza, 25200, Egypt
| | - Mubashir Hassan
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju-do 32588, South Korea
| | - Wael Mohamed
- Department of Clinical Pharmacology, Faculty of Medicine, Al-Menoufia University, Al-Menoufia, 25200 Egypt.,Basic Medical Science Department, Kulliyyah of Medicine, International Islamic University Malaysia, Kunatan Pahang, Malaysia
| | - Ahmed A Moustafa
- School of Social Sciences and Psychology and MARCS Institute for Brain and Behaviour, Western Sydney University, Sydney, New South Wales, 2751 Australia
| | - Mohamed Salama
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Al-Mansoura University, Al-Mansoura, 35111, Egypt.,Medical Experimental Research Center (MERC), Al-Mansoura University, Al-Mansoura, Egypt
| | - Dina Johar
- Department of Biochemistry and Nutrition, Faculty of Women for Arts, Sciences and Education, Ain Shams University, Heliopolis, Cairo, 11291, Egypt.,Max Rady College of Medicine, Rady Faculty of Health Sciences, Department of Physiology & Pathophysiology 432 Basic Medical Sciences Building, 745 Bannatyne Avenue University of Manitoba, Winnipeg, MB R3E 0J9, Canada, e-mail:
| | - Larry Bernstein
- Triplex Consulting, 54 Firethorn Lane, Northampton, MA 01060, USA
| |
Collapse
|
57
|
Transcriptome-pathology correlation identifies interplay between TDP-43 and the expression of its kinase CK1E in sporadic ALS. Acta Neuropathol 2018; 136:405-423. [PMID: 29881994 PMCID: PMC6215775 DOI: 10.1007/s00401-018-1870-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/18/2018] [Accepted: 05/23/2018] [Indexed: 12/12/2022]
Abstract
Sporadic amyotrophic lateral sclerosis (sALS) is the most common form of ALS, however, the molecular mechanisms underlying cellular damage and motor neuron degeneration remain elusive. To identify molecular signatures of sALS we performed genome-wide expression profiling in laser capture microdissection-enriched surviving motor neurons (MNs) from lumbar spinal cords of sALS patients with rostral onset and caudal progression. After correcting for immunological background, we discover a highly specific gene expression signature for sALS that is associated with phosphorylated TDP-43 (pTDP-43) pathology. Transcriptome–pathology correlation identified casein kinase 1ε (CSNK1E) mRNA as tightly correlated to levels of pTDP-43 in sALS patients. Enhanced crosslinking and immunoprecipitation in human sALS patient- and healthy control-derived frontal cortex, revealed that TDP-43 binds directly to and regulates the expression of CSNK1E mRNA. Additionally, we were able to show that pTDP-43 itself binds RNA. CK1E, the protein product of CSNK1E, in turn interacts with TDP-43 and promotes cytoplasmic accumulation of pTDP-43 in human stem-cell-derived MNs. Pathological TDP-43 phosphorylation is therefore, reciprocally regulated by CK1E activity and TDP-43 RNA binding. Our framework of transcriptome–pathology correlations identifies candidate genes with relevance to novel mechanisms of neurodegeneration.
Collapse
|
58
|
|
59
|
Lema YY, Gamo NJ, Yang K, Ishizuka K. Trait and state biomarkers for psychiatric disorders: Importance of infrastructure to bridge the gap between basic and clinical research and industry. Psychiatry Clin Neurosci 2018; 72:482-489. [PMID: 29687938 DOI: 10.1111/pcn.12669] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/18/2018] [Indexed: 12/30/2022]
Abstract
To further improve clinical activities in psychiatry by early diagnosis and early intervention with novel mechanism-guided treatments, there is a great need for biomarkers that reflect 'trait' and 'state' in major mental disorders. Stable trait biomarkers would allow early diagnosis, prognosis, and hopefully early intervention in these disorders, while dynamic state markers that reflect symptomatic changes would help to develop treatments that target these molecular mechanisms. However, in the search for such biomarkers, and eventually translating them to the clinic and industry, challenges currently exist at multiple levels, from basic scientific understanding, patient sample collection, and sample and data management, to bridging the gap between basic and clinical research and industry. To address these challenges, we propose an infrastructure that emphasizes: (i) a research and educational framework to facilitate translation between basic neuroscience, clinical research, and industry; (ii) patient recruitment and collection of disease-relevant samples to study trait and state biomarkers; and (iii) a comprehensive database to integrate patient and sample information with biological and clinical data. We believe that such an approach would bolster: research into the biological mechanisms of psychiatric disorders; and collaboration among the laboratory, clinic, and industry to translate these findings into successful treatments.
Collapse
Affiliation(s)
- Yukiko Y Lema
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Nao J Gamo
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Kun Yang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Koko Ishizuka
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
60
|
Whole transcriptome profiling of Late-Onset Alzheimer's Disease patients provides insights into the molecular changes involved in the disease. Sci Rep 2018. [PMID: 29523845 PMCID: PMC5844946 DOI: 10.1038/s41598-018-22701-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s Disease (AD) is the most common cause of dementia affecting the elderly population worldwide. We have performed a comprehensive transcriptome profiling of Late-Onset AD (LOAD) patients using second generation sequencing technologies, identifying 2,064 genes, 47 lncRNAs and 4 miRNAs whose expression is specifically deregulated in the hippocampal region of LOAD patients. Moreover, analyzing the hippocampal, temporal and frontal regions from the same LOAD patients, we identify specific sets of deregulated miRNAs for each region, and we confirm that the miR-132/212 cluster is deregulated in each of these regions in LOAD patients, consistent with these miRNAs playing a role in AD pathogenesis. Notably, a luciferase assay indicates that miR-184 is able to target the 3’UTR NR4A2 - which is known to be involved in cognitive functions and long-term memory and whose expression levels are inversely correlated with those of miR-184 in the hippocampus. Finally, RNA editing analysis reveals a general RNA editing decrease in LOAD hippocampus, with 14 recoding sites significantly and differentially edited in 11 genes. Our data underline specific transcriptional changes in LOAD brain and provide an important source of information for understanding the molecular changes characterizing LOAD progression.
Collapse
|
61
|
Benefit of Oleuropein Aglycone for Alzheimer's Disease by Promoting Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:5010741. [PMID: 29675133 PMCID: PMC5838478 DOI: 10.1155/2018/5010741] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 01/21/2018] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease is a proteinopathy characterized by accumulation of hyperphosphorylated Tau and β-amyloid. Autophagy is a physiological process by which aggregated proteins and damaged organelles are eliminated through lysosomal digestion. Autophagy deficiency has been demonstrated in Alzheimer's patients impairing effective elimination of aggregates and damaged mitochondria, leading to their accumulation, increasing their toxicity and oxidative stress. In the present study, we demonstrated by microarray analysis the downregulation of fundamental autophagy and mitophagy pathways in Alzheimer's patients. The benefits of the Mediterranean diet on Alzheimer's disease and cognitive impairment are well known, attributing this effect to several polyphenols, such as oleuropein aglycone (OLE), present in extra virgin olive oil. OLE is able to induce autophagy, achieving a decrease of aggregated proteins and a reduction of cognitive impairment in vivo. This effect is caused by the modulation of several pathways including the AMPK/mTOR axis and the activation of autophagy gene expression mediated by sirtuins and histone acetylation or EB transcription factor. We propose that supplementation of diet with extra virgin olive oil might have potential benefits for Alzheimer's patients by the induction of autophagy by OLE.
Collapse
|
62
|
Ramachandran S, Coffin SL, Tang TY, Jobaliya CD, Spengler RM, Davidson BL. Cis-acting single nucleotide polymorphisms alter MicroRNA-mediated regulation of human brain-expressed transcripts. Hum Mol Genet 2018; 25:4939-4950. [PMID: 28171541 PMCID: PMC5418741 DOI: 10.1093/hmg/ddw317] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/06/2016] [Accepted: 09/12/2016] [Indexed: 12/11/2022] Open
Abstract
Substantial variability exists in the presentation of complex neurological disorders, and the study of single nucleotide polymorphisms (SNPs) has shed light on disease mechanisms and pathophysiological variability in some cases. However, the vast majority of disease-linked SNPs have unidentified pathophysiological relevance. Here, we tested the hypothesis that SNPs within the miRNA recognition element (MRE; the region of the target transcript to which the miRNA binds) can impart changes in the expression of those genes, either by enhancing or reducing transcript and protein levels. To test this, we cross-referenced 7,153 miRNA-MRE brain interactions with the SNP database (dbSNP) to identify candidates, and functionally assessed 24 SNPs located in the 3’UTR or the coding sequence (CDS) of targets. For over half of the candidates tested, SNPs either enhanced (4 genes) or disrupted (10 genes) miRNA binding and target regulation. Additionally, SNPs causing a shift from a common to rare codon within the CDS facilitated miRNA binding downstream of the SNP, dramatically repressing target gene expression. The biological activity of the SNPs on miRNA regulation was also confirmed in induced pluripotent stem cell (iPSC) lines. These studies strongly support the notion that SNPs in the 3’UTR or the coding sequence of disease-relevant genes may be important in disease pathogenesis and should be reconsidered as candidate modifiers.
Collapse
Affiliation(s)
- Shyam Ramachandran
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Stephanie L Coffin
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Tin-Yun Tang
- Howard Hughes Medical Institute Medical Research Fellow, Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Chintan D Jobaliya
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, USA.,Human Pluripotent Stem Cell Core, Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ryan M Spengler
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Beverly L Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, USA.,The Department of Pathology & Laboratory Medicine, The Children’s Hospital of Philadelphia and The University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
63
|
Borrageiro G, Haylett W, Seedat S, Kuivaniemi H, Bardien S. A review of genome-wide transcriptomics studies in Parkinson's disease. Eur J Neurosci 2017; 47:1-16. [DOI: 10.1111/ejn.13760] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/26/2017] [Accepted: 10/19/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Genevie Borrageiro
- Division of Molecular Biology and Human Genetics; Department of Biomedical Sciences; Faculty of Medicine and Health Sciences; Stellenbosch University; PO Box 241 Cape Town South Africa
| | - William Haylett
- Division of Molecular Biology and Human Genetics; Department of Biomedical Sciences; Faculty of Medicine and Health Sciences; Stellenbosch University; PO Box 241 Cape Town South Africa
| | - Soraya Seedat
- Department of Psychiatry; Faculty of Medicine and Health Sciences; Stellenbosch University; Cape Town South Africa
| | - Helena Kuivaniemi
- Division of Molecular Biology and Human Genetics; Department of Biomedical Sciences; Faculty of Medicine and Health Sciences; Stellenbosch University; PO Box 241 Cape Town South Africa
- Department of Psychiatry; Faculty of Medicine and Health Sciences; Stellenbosch University; Cape Town South Africa
| | - Soraya Bardien
- Division of Molecular Biology and Human Genetics; Department of Biomedical Sciences; Faculty of Medicine and Health Sciences; Stellenbosch University; PO Box 241 Cape Town South Africa
| |
Collapse
|
64
|
Allen M, Wang X, Burgess JD, Watzlawik J, Serie DJ, Younkin CS, Nguyen T, Malphrus KG, Lincoln S, Carrasquillo MM, Ho C, Chakrabarty P, Strickland S, Murray ME, Swarup V, Geschwind DH, Seyfried NT, Dammer EB, Lah JJ, Levey AI, Golde TE, Funk C, Li H, Price ND, Petersen RC, Graff-Radford NR, Younkin SG, Dickson DW, Crook JR, Asmann YW, Ertekin-Taner N. Conserved brain myelination networks are altered in Alzheimer's and other neurodegenerative diseases. Alzheimers Dement 2017; 14:352-366. [PMID: 29107053 DOI: 10.1016/j.jalz.2017.09.012] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 09/13/2017] [Accepted: 09/20/2017] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Comparative transcriptome analyses in Alzheimer's disease (AD) and other neurodegenerative proteinopathies can uncover both shared and distinct disease pathways. METHODS We analyzed 940 brain transcriptomes including patients with AD, progressive supranuclear palsy (PSP; a primary tauopathy), and control subjects. RESULTS We identified transcriptional coexpression networks implicated in myelination, which were lower in PSP temporal cortex (TCX) compared with AD. Some of these associations were retained even after adjustments for brain cell population changes. These TCX myelination network structures were preserved in cerebellum but they were not differentially expressed in cerebellum between AD and PSP. Myelination networks were downregulated in both AD and PSP, when compared with control TCX samples. DISCUSSION Downregulation of myelination networks may underlie both PSP and AD pathophysiology, but may be more pronounced in PSP. These data also highlight conservation of transcriptional networks across brain regions and the influence of cell type changes on these networks.
Collapse
Affiliation(s)
- Mariet Allen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Xue Wang
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | | | - Jens Watzlawik
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Daniel J Serie
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - Curtis S Younkin
- Division of Information Technology, Mayo Clinic, Jacksonville, FL, USA
| | - Thuy Nguyen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Sarah Lincoln
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Charlotte Ho
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Paramita Chakrabarty
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | | | | | - Vivek Swarup
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Daniel H Geschwind
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA; Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric B Dammer
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - James J Lah
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Todd E Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Cory Funk
- Institute for Systems Biology, Seattle, WA, USA
| | - Hongdong Li
- Institute for Systems Biology, Seattle, WA, USA
| | | | | | | | | | | | - Julia R Crook
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - Yan W Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Department of Neurology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
65
|
Slower Dynamics and Aged Mitochondria in Sporadic Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9302761. [PMID: 29201274 PMCID: PMC5672147 DOI: 10.1155/2017/9302761] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/25/2017] [Accepted: 08/17/2017] [Indexed: 12/30/2022]
Abstract
Sporadic Alzheimer's disease corresponds to 95% of cases whose origin is multifactorial and elusive. Mitochondrial dysfunction is a major feature of Alzheimer's pathology, which might be one of the early events that trigger downstream principal events. Here, we show that multiple genes that control mitochondrial homeostasis, including fission and fusion, are downregulated in Alzheimer's patients. Additionally, we demonstrate that some of these dysregulations, such as diminished DLP1 levels and its mitochondrial localization, as well as reduced STOML2 and MFN2 fusion protein levels, take place in fibroblasts from sporadic Alzheimer's disease patients. The analysis of mitochondrial network disruption using CCCP indicates that the patients' fibroblasts exhibit slower dynamics and mitochondrial membrane potential recovery. These defects lead to strong accumulation of aged mitochondria in Alzheimer's fibroblasts. Accordingly, the analysis of autophagy and mitophagy involved genes in the patients demonstrates a downregulation indicating that the recycling mechanism of these aged mitochondria might be impaired. Our data reinforce the idea that mitochondrial dysfunction is one of the key early events of the disease intimately related with aging.
Collapse
|
66
|
Xu M, Zhang D, Luo R, Wu Y, Zhou H, Kong L, Bi R, Yao Y. A systematic integrated analysis of brain expression profiles reveals
YAP1
and other prioritized hub genes as important upstream regulators in Alzheimer's disease. Alzheimers Dement 2017; 14:215-229. [DOI: 10.1016/j.jalz.2017.08.012] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 08/06/2017] [Accepted: 08/07/2017] [Indexed: 01/28/2023]
Affiliation(s)
- Min Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province Kunming Institute of Zoology Kunming Yunnan China
- Kunming College of Life Science University of Chinese Academy of Sciences Kunming Yunnan China
| | - Deng‐Feng Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province Kunming Institute of Zoology Kunming Yunnan China
| | - Rongcan Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province Kunming Institute of Zoology Kunming Yunnan China
- Kunming College of Life Science University of Chinese Academy of Sciences Kunming Yunnan China
| | - Yong Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province Kunming Institute of Zoology Kunming Yunnan China
- Kunming College of Life Science University of Chinese Academy of Sciences Kunming Yunnan China
| | - Hejiang Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province Kunming Institute of Zoology Kunming Yunnan China
| | - Li‐Li Kong
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province Kunming Institute of Zoology Kunming Yunnan China
- Institute of Health Science Anhui University Hefei Anhui China
| | - Rui Bi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province Kunming Institute of Zoology Kunming Yunnan China
| | - Yong‐Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province Kunming Institute of Zoology Kunming Yunnan China
- Kunming College of Life Science University of Chinese Academy of Sciences Kunming Yunnan China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences Shanghai China
| |
Collapse
|
67
|
D'Erchia AM, Gallo A, Manzari C, Raho S, Horner DS, Chiara M, Valletti A, Aiello I, Mastropasqua F, Ciaccia L, Locatelli F, Pisani F, Nicchia GP, Svelto M, Pesole G, Picardi E. Massive transcriptome sequencing of human spinal cord tissues provides new insights into motor neuron degeneration in ALS. Sci Rep 2017; 7:10046. [PMID: 28855684 PMCID: PMC5577269 DOI: 10.1038/s41598-017-10488-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 08/09/2017] [Indexed: 12/13/2022] Open
Abstract
ALS is a devastating and debilitating human disease characterized by the progressive death of upper and lower motor neurons. Although much effort has been made to elucidate molecular determinants underlying the onset and progression of the disorder, the causes of ALS remain largely unknown. In the present work, we have deeply sequenced whole transcriptome from spinal cord ventral horns of post-mortem ALS human donors affected by the sporadic form of the disease (which comprises ~90% of the cases but which is less investigated than the inherited form of the disease). We observe 1160 deregulated genes including 18 miRNAs and show that down regulated genes are mainly of neuronal derivation while up regulated genes have glial origin and tend to be involved in neuroinflammation or cell death. Remarkably, we find strong deregulation of SNAP25 and STX1B at both mRNA and protein levels suggesting impaired synaptic function through SNAP25 reduction as a possible cause of calcium elevation and glutamate excitotoxicity. We also note aberrant alternative splicing but not disrupted RNA editing.
Collapse
Affiliation(s)
- Anna Maria D'Erchia
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70126, Bari, Italy.,Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Via Amendola 165/A, 70126, Bari, Italy
| | - Angela Gallo
- Department of Pediatric Oncohaematology, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Caterina Manzari
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Via Amendola 165/A, 70126, Bari, Italy
| | - Susanna Raho
- Department of Pediatric Oncohaematology, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - David S Horner
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Matteo Chiara
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Via Amendola 165/A, 70126, Bari, Italy
| | - Alessio Valletti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Via Amendola 165/A, 70126, Bari, Italy
| | - Italia Aiello
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Via Amendola 165/A, 70126, Bari, Italy
| | - Francesca Mastropasqua
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70126, Bari, Italy
| | - Loredana Ciaccia
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70126, Bari, Italy
| | - Franco Locatelli
- Department of Pediatric Oncohaematology, Bambino Gesù Children's Hospital IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Francesco Pisani
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70126, Bari, Italy
| | - Grazia Paola Nicchia
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70126, Bari, Italy
| | - Maria Svelto
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70126, Bari, Italy.,National Institute of Biostructures and Biosystems (INBB), Viale Medaglie D'Oro 305, 00136, Rome, Italy.,Center of Excellence in Comparative Genomics, University of Bari, Piazza Umberto I, 70121, Bari, Italy
| | - Graziano Pesole
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70126, Bari, Italy. .,Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Via Amendola 165/A, 70126, Bari, Italy. .,National Institute of Biostructures and Biosystems (INBB), Viale Medaglie D'Oro 305, 00136, Rome, Italy. .,Center of Excellence in Comparative Genomics, University of Bari, Piazza Umberto I, 70121, Bari, Italy.
| | - Ernesto Picardi
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Via Orabona 4, 70126, Bari, Italy. .,Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Via Amendola 165/A, 70126, Bari, Italy. .,National Institute of Biostructures and Biosystems (INBB), Viale Medaglie D'Oro 305, 00136, Rome, Italy.
| |
Collapse
|
68
|
Repulsive Guidance Molecule a (RGMa) Induces Neuropathological and Behavioral Changes That Closely Resemble Parkinson's Disease. J Neurosci 2017; 37:9361-9379. [PMID: 28842419 DOI: 10.1523/jneurosci.0084-17.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 07/12/2017] [Accepted: 08/11/2017] [Indexed: 01/06/2023] Open
Abstract
Repulsive guidance molecule member a (RGMa) is a membrane-associated or released guidance molecule that is involved in axon guidance, cell patterning, and cell survival. In our previous work, we showed that RGMa is significantly upregulated in the substantia nigra of patients with Parkinson's disease. Here we demonstrate the expression of RGMa in midbrain human dopaminergic (DA) neurons. To investigate whether RGMa might model aspects of the neuropathology of Parkinson's disease in mouse, we targeted RGMa to adult midbrain dopaminergic neurons using adeno-associated viral vectors. Overexpression of RGMa resulted in a progressive movement disorder, including motor coordination and imbalance, which is typical for a loss of DA release in the striatum. In line with this, RGMa induced selective degeneration of dopaminergic neurons in the substantia nigra (SN) and affected the integrity of the nigrostriatal system. The degeneration of dopaminergic neurons was accompanied by a strong microglia and astrocyte activation. The behavioral, molecular, and anatomical changes induced by RGMa in mice are remarkably similar to the clinical and neuropathological hallmarks of Parkinson's disease. Our data indicate that dysregulation of RGMa plays an important role in the pathology of Parkinson's disease, and antibody-mediated functional interference with RGMa may be a disease modifying treatment option.SIGNIFICANCE STATEMENT Parkinson's disease (PD) is a neurodegenerative disease characterized by severe motor dysfunction due to progressive degeneration of mesencephalic dopaminergic (DA) neurons in the substantia nigra. To date, there is no regenerative treatment available. We previously showed that repulsive guidance molecule member a (RGMa) is upregulated in the substantia nigra of PD patients. Adeno-associated virus-mediated targeting of RGMa to mouse DA neurons showed that overexpression of this repulsive axon guidance and cell patterning cue models the behavioral and neuropathological characteristics of PD in a remarkable way. These findings have implications for therapy development as interfering with the function of this specific axon guidance cue may be beneficial to the survival of DA neurons.
Collapse
|
69
|
Topa H, Honkela A. Analysis of differential splicing suggests different modes of short-term splicing regulation. Bioinformatics 2017; 32:i147-i155. [PMID: 27307611 PMCID: PMC4908367 DOI: 10.1093/bioinformatics/btw283] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Motivation: Alternative splicing is an important mechanism in which the regions of pre-mRNAs are differentially joined in order to form different transcript isoforms. Alternative splicing is involved in the regulation of normal physiological functions but also linked to the development of diseases such as cancer. We analyse differential expression and splicing using RNA-sequencing time series in three different settings: overall gene expression levels, absolute transcript expression levels and relative transcript expression levels. Results: Using estrogen receptor α signaling response as a model system, our Gaussian process-based test identifies genes with differential splicing and/or differentially expressed transcripts. We discover genes with consistent changes in alternative splicing independent of changes in absolute expression and genes where some transcripts change whereas others stay constant in absolute level. The results suggest classes of genes with different modes of alternative splicing regulation during the experiment. Availability and Implementation: R and Matlab codes implementing the method are available at https://github.com/PROBIC/diffsplicing. An interactive browser for viewing all model fits is available at http://users.ics.aalto.fi/hande/splicingGP/ Contact:hande.topa@helsinki.fi or antti.honkela@helsinki.fi Supplementary information:Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Hande Topa
- Helsinki Institute for Information Technology HIIT, Department of Computer Science, Aalto University, Espoo 00076, Finland Helsinki Institute for Information Technology HIIT, Department of Computer Science, University of Helsinki, Helsinki 00014, Finland
| | - Antti Honkela
- Helsinki Institute for Information Technology HIIT, Department of Computer Science, University of Helsinki, Helsinki 00014, Finland
| |
Collapse
|
70
|
Lall D, Baloh RH. Microglia and C9orf72 in neuroinflammation and ALS and frontotemporal dementia. J Clin Invest 2017; 127:3250-3258. [PMID: 28737506 DOI: 10.1172/jci90607] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a degenerative disorder that is characterized by loss of motor neurons and shows clinical, pathological, and genetic overlap with frontotemporal dementia (FTD). Activated microglia are a universal feature of ALS/FTD pathology; however, their role in disease pathogenesis remains incompletely understood. The recent discovery that ORF 72 on chromosome 9 (C9orf72), the gene most commonly mutated in ALS/FTD, has an important role in myeloid cells opened the possibility that altered microglial function plays an active role in disease. This Review highlights the contribution of microglia to ALS/FTD pathogenesis, discusses the connection between autoimmunity and ALS/FTD, and explores the possibility that C9orf72 and other ALS/FTD genes may have a "dual effect" on both neuronal and myeloid cell function that could explain a shared propensity for altered systemic immunity and neurodegeneration.
Collapse
Affiliation(s)
- Deepti Lall
- Board of Governors Regenerative Medicine Institute and
| | - Robert H Baloh
- Board of Governors Regenerative Medicine Institute and.,Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
71
|
Hautbergue GM, Castelli LM, Ferraiuolo L, Sanchez-Martinez A, Cooper-Knock J, Higginbottom A, Lin YH, Bauer CS, Dodd JE, Myszczynska MA, Alam SM, Garneret P, Chandran JS, Karyka E, Stopford MJ, Smith EF, Kirby J, Meyer K, Kaspar BK, Isaacs AM, El-Khamisy SF, De Vos KJ, Ning K, Azzouz M, Whitworth AJ, Shaw PJ. SRSF1-dependent nuclear export inhibition of C9ORF72 repeat transcripts prevents neurodegeneration and associated motor deficits. Nat Commun 2017; 8:16063. [PMID: 28677678 PMCID: PMC5504286 DOI: 10.1038/ncomms16063] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 05/24/2017] [Indexed: 12/13/2022] Open
Abstract
Hexanucleotide repeat expansions in the C9ORF72 gene are the commonest known genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Expression of repeat transcripts and dipeptide repeat proteins trigger multiple mechanisms of neurotoxicity. How repeat transcripts get exported from the nucleus is unknown. Here, we show that depletion of the nuclear export adaptor SRSF1 prevents neurodegeneration and locomotor deficits in a Drosophila model of C9ORF72-related disease. This intervention suppresses cell death of patient-derived motor neuron and astrocytic-mediated neurotoxicity in co-culture assays. We further demonstrate that either depleting SRSF1 or preventing its interaction with NXF1 specifically inhibits the nuclear export of pathological C9ORF72 transcripts, the production of dipeptide-repeat proteins and alleviates neurotoxicity in Drosophila, patient-derived neurons and neuronal cell models. Taken together, we show that repeat RNA-sequestration of SRSF1 triggers the NXF1-dependent nuclear export of C9ORF72 transcripts retaining expanded hexanucleotide repeats and reveal a novel promising therapeutic target for neuroprotection.
Collapse
Affiliation(s)
- Guillaume M. Hautbergue
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Lydia M. Castelli
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Alvaro Sanchez-Martinez
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Ya-Hui Lin
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Claudia S. Bauer
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Jennifer E. Dodd
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Monika A. Myszczynska
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Sarah M. Alam
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Pierre Garneret
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Jayanth S. Chandran
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Evangelia Karyka
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Matthew J. Stopford
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Emma F. Smith
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Janine Kirby
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Kathrin Meyer
- Nationwide Children’s Research Institute, Department of Pediatrics, The Ohio State University, 700 Children’s Drive, Rm. WA3022, Columbus, Ohio 43205, USA
| | - Brian K. Kaspar
- Nationwide Children’s Research Institute, Department of Pediatrics, The Ohio State University, 700 Children’s Drive, Rm. WA3022, Columbus, Ohio 43205, USA
| | - Adrian M. Isaacs
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Sherif F. El-Khamisy
- Krebs Institute, Department of Molecular Biology and Biotechnology, University of Sheffield, Firth Court, Sheffield S10 2TN, UK
| | - Kurt J. De Vos
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Ke Ning
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Mimoun Azzouz
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| | - Alexander J. Whitworth
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield S10 2HQ, UK
| |
Collapse
|
72
|
Abstract
Three neurodegenerative diseases [Amyotrophic Lateral Sclerosis (ALS), Parkinson's disease (PD) and Alzheimer's disease (AD)] have many characteristics like pathological mechanisms and genes. In this sense some researchers postulate that these diseases share the same alterations and that one alteration in a specific protein triggers one of these diseases. Analyses of gene expression may shed more light on how to discover pathways, pathologic mechanisms associated with the disease, biomarkers and potential therapeutic targets. In this review, we analyze four microarrays related to three neurodegenerative diseases. We will systematically examine seven genes (CHN1, MDH1, PCP4, RTN1, SLC14A1, SNAP25 and VSNL1) that are altered in the three neurodegenerative diseases. A network was built and used to identify pathways, miRNA and drugs associated with ALS, AD and PD using Cytoscape software an interaction network based on the protein interactions of these genes. The most important affected pathway is PI3K-Akt signalling. Thirteen microRNAs (miRNA-19B1, miRNA-107, miRNA-124-1, miRNA-124-2, miRNA-9-2, miRNA-29A, miRNA-9-3, miRNA-328, miRNA-19B2, miRNA-29B2, miRNA-124-3, miRNA-15A and miRNA-9-1) and four drugs (Estradiol, Acetaminophen, Resveratrol and Progesterone) for new possible treatments were identified.
Collapse
Affiliation(s)
| | - Marcelo Alarcón
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile; Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca 3460000, Chile.
| |
Collapse
|
73
|
Lachen-Montes M, Zelaya MV, Segura V, Fernández-Irigoyen J, Santamaría E. Progressive modulation of the human olfactory bulb transcriptome during Alzheimer´s disease evolution: novel insights into the olfactory signaling across proteinopathies. Oncotarget 2017; 8:69663-69679. [PMID: 29050232 PMCID: PMC5642507 DOI: 10.18632/oncotarget.18193] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 05/07/2017] [Indexed: 01/01/2023] Open
Abstract
Alzheimer´s disease (AD) is characterized by progressive dementia, initially presenting olfactory dysfunction. Despite the olfactory bulb (OB) is the first central structure of the olfactory pathway, we lack a complete molecular characterization of the transcriptional events that occurs in this olfactory area during AD progression. To address this gap in knowledge, we have assessed the genome-wide expression in postmortem OBs from subjects with varying degree of AD pathology. A stage-dependent deregulation of specific pathways was observed, revealing transmembrane transport, and neuroinflammation as part of the functional modules that are disrupted across AD grading. Potential drivers of neurodegeneration predicted by network-driven transcriptomics were monitored across different types of dementia, including progressive supranuclear palsy (PSP), mixed dementia, and frontotemporal lobar degeneration (FTLD). Epidermal growth factor receptor (EGFR) expression was significantly increased in the OB of AD and mixed dementia subjects. Moreover, a significant increment in the activation of signal transducer and activator of transcription 3 (STAT3) was exclusively detected in advanced AD stages, whereas total STAT3 levels were specifically overexpressed in mixed dementia. Furthermore, transcription factors deregulated in the OB of mixed dementia subjects such as cAMP Responsive Element Binding Protein 1 (CREB1) and AP-1 Transcription Factor Subunit (c-Jun) were not differentially modulated at olfactory level across AD grading. On the other hand, olfactory expression of this signal transducer panel was unchanged in PSP and FTLD subjects. Taken together, this study unveils cross-disease similarities and differences for specific signal transducers, providing mechanistic clues to the intriguing divergence of AD pathology across proteinopathies.
Collapse
Affiliation(s)
- Mercedes Lachen-Montes
- Clinical Neuroproteomics Group, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain
| | - María Victoria Zelaya
- Clinical Neuroproteomics Group, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Pathological Anatomy Department, Navarra Hospital Complex, Pamplona, Spain
| | - Víctor Segura
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Bioinformatics Unit, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Group, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Group, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain
| |
Collapse
|
74
|
Gama-Carvalho M, L Garcia-Vaquero M, R Pinto F, Besse F, Weis J, Voigt A, Schulz JB, De Las Rivas J. Linking amyotrophic lateral sclerosis and spinal muscular atrophy through RNA-transcriptome homeostasis: a genomics perspective. J Neurochem 2017; 141:12-30. [PMID: 28054357 DOI: 10.1111/jnc.13945] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/02/2016] [Accepted: 12/24/2016] [Indexed: 12/11/2022]
Abstract
In this review, we present our most recent understanding of key biomolecular processes that underlie two motor neuron degenerative disorders, amyotrophic lateral sclerosis, and spinal muscular atrophy. We focus on the role of four multifunctional proteins involved in RNA metabolism (TDP-43, FUS, SMN, and Senataxin) that play a causal role in these diseases. Recent results have led to a novel scenario of intricate connections between these four proteins, bringing transcriptome homeostasis into the spotlight as a common theme in motor neuron degeneration. We review reported functional and physical interactions between these four proteins, highlighting their common association with nuclear bodies and small nuclear ribonucleoprotein particle biogenesis and function. We discuss how these interactions are turning out to be particularly relevant for the control of transcription and chromatin homeostasis, including the recent identification of an association between SMN and Senataxin required to ensure the resolution of DNA-RNA hybrid formation and proper termination by RNA polymerase II. These connections strongly support the existence of common pathways underlying the spinal muscular atrophy and amyotrophic lateral sclerosis phenotype. We also discuss the potential of genome-wide expression profiling, in particular RNA sequencing derived data, to contribute to unravelling the underlying mechanisms. We provide a review of publicly available datasets that have addressed both diseases using these approaches, and highlight the value of investing in cross-disease studies to promote our understanding of the pathways leading to neurodegeneration.
Collapse
Affiliation(s)
- Margarida Gama-Carvalho
- Universidade de Lisboa, Faculdade de Ciências, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, 1749-016 Lisboa, Portugal
| | - Marina L Garcia-Vaquero
- Universidade de Lisboa, Faculdade de Ciências, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, 1749-016 Lisboa, Portugal
| | - Francisco R Pinto
- Universidade de Lisboa, Faculdade de Ciências, BioISI - Biosystems & Integrative Sciences Institute, Campo Grande, 1749-016 Lisboa, Portugal
| | | | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University, Aachen, Germany
| | - Aaron Voigt
- Department of Neurology, University Hospital, RWTH Aachen University, Aachen, Germany.,JARA-Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| | - Jörg B Schulz
- Department of Neurology, University Hospital, RWTH Aachen University, Aachen, Germany.,JARA-Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| | - Javier De Las Rivas
- Cancer Research Center (CiC-IBMCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca (USAL), Salamanca, Spain
| |
Collapse
|
75
|
Pharmacological intervention of early neuropathy in neurodegenerative diseases. Pharmacol Res 2017; 119:169-177. [PMID: 28167240 DOI: 10.1016/j.phrs.2017.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/31/2017] [Accepted: 02/02/2017] [Indexed: 12/11/2022]
Abstract
Extensive studies have reported the significant roles of numerous cellular features and processes in properly maintaining neuronal morphology and function throughout the lifespan of an animal. Any alterations in their homeostasis appear to be strongly associated with neuronal aging and the pathogenesis of various neurodegenerative diseases, even before the occurrence of prominent neuronal death. However, until recently, the primary focus of studies regarding many neurodegenerative diseases has been on the massive cell death occurring at the late stages of disease progression. Thus, our understanding on early neuropathy in these diseases remains relatively limited. The complicated nature of various neuropathic features manifested early in neurodegenerative diseases suggests the involvement of a system-wide transcriptional regulation and epigenetic control. Epigenetic alterations and consequent changes in the neuronal transcriptome are now begun to be extensively studied in various neurodegenerative diseases. Upon the catastrophic incident of neuronal death in disease progression, it is utterly difficult to reverse the deleterious defects by pharmacological treatments, and therefore, therapeutics targeting the system-wide transcriptional dysregulation associated with specific early neuropathy is considered a better option. Here, we review our current understanding on the system-wide transcriptional dysregulation that is likely associated with early neuropathy shown in various neurodegenerative diseases and discuss the possible future developments of pharmaceutical therapeutics.
Collapse
|
76
|
Identifying a gene expression signature of cluster headache in blood. Sci Rep 2017; 7:40218. [PMID: 28074859 PMCID: PMC5225606 DOI: 10.1038/srep40218] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 12/05/2016] [Indexed: 12/12/2022] Open
Abstract
Cluster headache is a relatively rare headache disorder, typically characterized by multiple daily, short-lasting attacks of excruciating, unilateral (peri-)orbital or temporal pain associated with autonomic symptoms and restlessness. To better understand the pathophysiology of cluster headache, we used RNA sequencing to identify differentially expressed genes and pathways in whole blood of patients with episodic (n = 19) or chronic (n = 20) cluster headache in comparison with headache-free controls (n = 20). Gene expression data were analysed by gene and by module of co-expressed genes with particular attention to previously implicated disease pathways including hypocretin dysregulation. Only moderate gene expression differences were identified and no associations were found with previously reported pathogenic mechanisms. At the level of functional gene sets, associations were observed for genes involved in several brain-related mechanisms such as GABA receptor function and voltage-gated channels. In addition, genes and modules of co-expressed genes showed a role for intracellular signalling cascades, mitochondria and inflammation. Although larger study samples may be required to identify the full range of involved pathways, these results indicate a role for mitochondria, intracellular signalling and inflammation in cluster headache.
Collapse
|
77
|
Qin DP, Feng XL, Zhang WY, Gao H, Cheng XR, Zhou WX, Yu Y, Yao XS. Anti-neuroinflammatory asarone derivatives from the rhizomes of Acorus tatarinowii. RSC Adv 2017. [DOI: 10.1039/c6ra27786a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A novel 7-O-7′ type mesomeric neolignan,meso-asarolignan A (1), six pairs of new neolignan enantiomers, (±)-asarolignan B–G (2a/2b,3a/3b,4–6,7a/7b), along with 16 known analogues (8–23) were isolated from the rhizomes ofAcorus tatarinowii.
Collapse
Affiliation(s)
- Da-Peng Qin
- Institute of Traditional Chinese Medicine & Natural Products
- College of Pharmacy
- Jinan University
- Guangzhou 510632
- P. R. China
| | - Xiao-Lin Feng
- Institute of Traditional Chinese Medicine & Natural Products
- College of Pharmacy
- Jinan University
- Guangzhou 510632
- P. R. China
| | - Wei-Yang Zhang
- State Key Laboratory of Quality Research in Chinese Medicine
- Macau University of Science and Technology
- Macau
- P. R. China
| | - Hao Gao
- Institute of Traditional Chinese Medicine & Natural Products
- College of Pharmacy
- Jinan University
- Guangzhou 510632
- P. R. China
| | - Xiao-Rui Cheng
- Beijing Institute of Pharmacology and Toxicology
- Beijing 100850
- P. R. China
| | - Wen-Xia Zhou
- Beijing Institute of Pharmacology and Toxicology
- Beijing 100850
- P. R. China
| | - Yang Yu
- Institute of Traditional Chinese Medicine & Natural Products
- College of Pharmacy
- Jinan University
- Guangzhou 510632
- P. R. China
| | - Xin-Sheng Yao
- Institute of Traditional Chinese Medicine & Natural Products
- College of Pharmacy
- Jinan University
- Guangzhou 510632
- P. R. China
| |
Collapse
|
78
|
John S, Mishra R. mRNA Transcriptomics of Galectins Unveils Heterogeneous Organization in Mouse and Human Brain. Front Mol Neurosci 2016; 9:139. [PMID: 28018170 PMCID: PMC5159438 DOI: 10.3389/fnmol.2016.00139] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 11/23/2016] [Indexed: 12/22/2022] Open
Abstract
Background: Galectins, a family of non-classically secreted, β-galactoside binding proteins is involved in several brain disorders; however, no systematic knowledge on the normal neuroanatomical distribution and functions of galectins exits. Hence, the major purpose of this study was to understand spatial distribution and predict functions of galectins in brain and also compare the degree of conservation vs. divergence between mouse and human species. The latter objective was required to determine the relevance and appropriateness of studying galectins in mouse brain which may ultimately enable us to extrapolate the findings to human brain physiology and pathologies. Results: In order to fill this crucial gap in our understanding of brain galectins, we analyzed the in situ hybridization and microarray data of adult mouse and human brain respectively, from the Allen Brain Atlas, to resolve each galectin-subtype’s spatial distribution across brain distinct cytoarchitecture. Next, transcription factors (TFs) that may regulate galectins were identified using TRANSFAC software and the list obtained was further curated to sort TFs on their confirmed transcript expression in the adult brain. Galectin-TF cluster analysis, gene-ontology annotations and co-expression networks were then extrapolated to predict distinct functional relevance of each galectin in the neuronal processes. Data shows that galectins have highly heterogeneous expression within and across brain sub-structures and are predicted to be the crucial targets of brain enriched TFs. Lgals9 had maximal spatial distribution across mouse brain with inferred predominant roles in neurogenesis while LGALS1 was ubiquitously expressed in human. Limbic region associated with learning, memory and emotions and substantia nigra associated with motor movements showed strikingly high expression of LGALS1 and LGALS8 in human vs. mouse brain. The overall expression profile of galectin-8 was most preserved across both these species, however, galectin-9 showed maximal preservation only in the cerebral cortex. Conclusion: It is for the first time that a comprehensive description of galectins’ mRNA expression profile in brain is presented. Results suggests that spatial transcriptome changes in galectins may contribute to differential brain functions and evolution across species that highlights galectins as novel signatures of brain heterogeneity and functions, which if disturbed, can promote several brain disorders.
Collapse
Affiliation(s)
- Sebastian John
- Disease Biology Program, Department of Neurobiology and Genetics, Rajiv Gandhi Centre for Biotechnology Thiruvananthapuram, India
| | - Rashmi Mishra
- Disease Biology Program, Department of Neurobiology and Genetics, Rajiv Gandhi Centre for Biotechnology Thiruvananthapuram, India
| |
Collapse
|
79
|
Riva N, Clarelli F, Domi T, Cerri F, Gallia F, Trimarco A, Brambilla P, Lunetta C, Lazzerini A, Lauria G, Taveggia C, Iannaccone S, Nobile-Orazio E, Comi G, D’Antonio M, Martinelli-Boneschi F, Quattrini A. Unraveling gene expression profiles in peripheral motor nerve from amyotrophic lateral sclerosis patients: insights into pathogenesis. Sci Rep 2016; 6:39297. [PMID: 27982123 PMCID: PMC5159906 DOI: 10.1038/srep39297] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 11/21/2016] [Indexed: 01/12/2023] Open
Abstract
The aim of the present study is to investigate the molecular pathways underlying amyotrophic lateral sclerosis (ALS) pathogenesis within the peripheral nervous system. We analyzed gene expression changes in human motor nerve diagnostic biopsies obtained from eight ALS patients and seven patients affected by motor neuropathy as controls. An integrated transcriptomics and system biology approach was employed. We identified alterations in the expression of 815 genes, with 529 up-regulated and 286 down-regulated in ALS patients. Up-regulated genes clustered around biological process involving RNA processing and protein metabolisms. We observed a significant enrichment of up-regulated small nucleolar RNA transcripts (p = 2.68*10-11) and genes related to endoplasmic reticulum unfolded protein response and chaperone activity. We found a significant down-regulation in ALS of genes related to the glutamate metabolism. Interestingly, a network analysis highlighted HDAC2, belonging to the histone deacetylase family, as the most interacting node. While so far gene expression studies in human ALS have been performed in postmortem tissues, here specimens were obtained from biopsy at an early phase of the disease, making these results new in the field of ALS research and therefore appealing for gene discovery studies.
Collapse
Affiliation(s)
- Nilo Riva
- Experimental Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Ferdinando Clarelli
- Laboratory of Genetics of Complex Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Teuta Domi
- Experimental Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Federica Cerri
- Experimental Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Gallia
- 2Neurology, IRCCS Istituto Clinico Humanitas, Milano University, Milan, Italy
| | - Amelia Trimarco
- Axo-glia interactions Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Paola Brambilla
- Laboratory of Genetics of Complex Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Christian Lunetta
- NEuroMuscular Omnicentre (NEMO), Niguarda Ca Granda Hospital, Milan, Italy
| | - Alberto Lazzerini
- Hand Surgery and Microsurgery Unit, IRCCS Humanitas Clinical Institute, Milan, Italy
| | - Giuseppe Lauria
- 3rd Neurology Unit, IRCCS Foundation “Carlo Besta” Neurological Institute, Milan, Italy
| | - Carla Taveggia
- Axo-glia interactions Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Sandro Iannaccone
- Department of Clinical Neurosciences, San Raffaele Scientific Institute, Milan, Italy
| | | | - Giancarlo Comi
- Experimental Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
- Universita` Vita e Salute San Raffaele, Milan, Italy
| | - Maurizio D’Antonio
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Filippo Martinelli-Boneschi
- Laboratory of Genetics of Complex Neurological Disorders, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Angelo Quattrini
- Experimental Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
80
|
Solaimani P, Saffari A, Sioutas C, Bondy SC, Campbell A. Exposure to ambient ultrafine particulate matter alters the expression of genes in primary human neurons. Neurotoxicology 2016; 58:50-57. [PMID: 27851901 DOI: 10.1016/j.neuro.2016.11.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/07/2016] [Accepted: 11/08/2016] [Indexed: 11/18/2022]
Abstract
Exposure to ambient particulate matter (PM) has been associated with the onset of neurodevelopmental and neurodegenerative disorders, but the mechanism of toxicity remains unclear. To gain insight into this neurotoxicity, this study sought to examine global gene expression changes caused by exposure to ambient ultrafine PM. Microarray analysis was performed on primary human neurons derived from fetal brain tissue after a 24h exposure to 20μg/mL of ambient ultrafine particles. We found a majority of the changes in noncoding RNAs, which are involved in epigenetic regulation of gene expression, and thereby could impact the expression of several other protein coding gene targets. Although neurons from biologically different lot numbers were used, we found a significant increase in the expression of metallothionein 1A and 1F in all samples after exposure to particulate matter as confirmed by quantitative PCR. These metallothionein 1 proteins are responsible for neuroprotection after exposure to environmental insult but prolonged induction can be toxic. Epidemiological studies have reported that in utero exposure to ultrafine PM not only leads to neurodevelopmental and behavioral abnormalities, but may also predispose the progeny to neurodegenerative disease later in life by genetic imprinting. Our results pinpoint some of the PM-induced genetic changes that may underlie these findings.
Collapse
Affiliation(s)
- Parrisa Solaimani
- Department of Pharmaceutical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Arian Saffari
- Department of Civil and Environmental Engineering, University of Southern California, Los Angeles, CA, USA
| | - Constantinos Sioutas
- Department of Civil and Environmental Engineering, University of Southern California, Los Angeles, CA, USA
| | - Stephen C Bondy
- Center for Occupational and Environmental Health, Department of Medicine, University of California, Irvine, CA, USA
| | - Arezoo Campbell
- Department of Pharmaceutical Sciences, Western University of Health Sciences, Pomona, CA, USA.
| |
Collapse
|
81
|
Voyle N, Keohane A, Newhouse S, Lunnon K, Johnston C, Soininen H, Kloszewska I, Mecocci P, Tsolaki M, Vellas B, Lovestone S, Hodges A, Kiddle S, Dobson RJ. A Pathway Based Classification Method for Analyzing Gene Expression for Alzheimer's Disease Diagnosis. J Alzheimers Dis 2016; 49:659-69. [PMID: 26484910 PMCID: PMC4927941 DOI: 10.3233/jad-150440] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background: Recent studies indicate that gene expression levels in blood may be able to differentiate subjects with Alzheimer’s disease (AD) from normal elderly controls and mild cognitively impaired (MCI) subjects. However, there is limited replicability at the single marker level. A pathway-based interpretation of gene expression may prove more robust. Objectives: This study aimed to investigate whether a case/control classification model built on pathway level data was more robust than a gene level model and may consequently perform better in test data. The study used two batches of gene expression data from the AddNeuroMed (ANM) and Dementia Case Registry (DCR) cohorts. Methods: Our study used Illumina Human HT-12 Expression BeadChips to collect gene expression from blood samples. Random forest modeling with recursive feature elimination was used to predict case/control status. Age and APOE ɛ4 status were used as covariates for all analysis. Results: Gene and pathway level models performed similarly to each other and to a model based on demographic information only. Conclusions: Any potential increase in concordance from the novel pathway level approach used here has not lead to a greater predictive ability in these datasets. However, we have only tested one method for creating pathway level scores. Further, we have been able to benchmark pathways against genes in datasets that had been extensively harmonized. Further work should focus on the use of alternative methods for creating pathway level scores, in particular those that incorporate pathway topology, and the use of an endophenotype based approach.
Collapse
Affiliation(s)
- Nicola Voyle
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,MRC Social, Genetic and Developmental Psychiatry Centre, King's College London, London, UK
| | - Aoife Keohane
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Stephen Newhouse
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | | | - Caroline Johnston
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Hilkka Soininen
- Department of Neurology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | | | - Patrizia Mecocci
- Institute of Gerontology and Geriatrics, University of Perugia, Perugia, Italy
| | - Magda Tsolaki
- 3rd Department of Neurology, Aristotle University, Thessaloniki, Greece
| | | | - Simon Lovestone
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,Department of Pyschiatry, Oxford University, Oxford, UK
| | - Angela Hodges
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Steven Kiddle
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,MRC Social, Genetic and Developmental Psychiatry Centre, King's College London, London, UK
| | - Richard Jb Dobson
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| |
Collapse
|
82
|
An Intrabody Drug (rAAV6-INT41) Reduces the Binding of N-Terminal Huntingtin Fragment(s) to DNA to Basal Levels in PC12 Cells and Delays Cognitive Loss in the R6/2 Animal Model. JOURNAL OF NEURODEGENERATIVE DISEASES 2016; 2016:7120753. [PMID: 27595037 PMCID: PMC4995342 DOI: 10.1155/2016/7120753] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 06/27/2016] [Indexed: 01/01/2023]
Abstract
Huntington's disease (HD) is a fatal progressive disease linked to expansion of glutamine repeats in the huntingtin protein and characterized by the progressive loss of cognitive and motor function. We show that expression of a mutant human huntingtin exon-1-GFP fusion construct results in nonspecific gene dysregulation that is significantly reduced by 50% due to coexpression of INT41, an intrabody specific for the proline-rich region of the huntingtin protein. Using stable PC12 cell lines expressing either inducible human mutant huntingtin (mHtt, Q73) or normal huntingtin (nHtt, Q23), we investigated the effect of rAAV6-INT41, an adeno-associated virus vector with the INT41 coding sequence, on the subcellular distribution of Htt. Compartmental fractionation 8 days after induction of Htt showed a 6-fold increased association of a dominate N-terminal mHtt fragment with DNA compared to N-terminal nHtt. Transduction with rAAV6-INT41 reduced DNA binding of N-terminal mHtt 6.5-fold in the nucleus and reduced nuclear translocation of the detected fragments. Subsequently, when rAAV6-INT41 is delivered to the striatum in the R6/2 mouse model, treated female mice exhibited executive function statistically indistinguishable from wild type, accompanied by reductions in Htt aggregates in the striatum, suggesting that rAAV6-INT41 is promising as a gene therapy for Huntington's disease.
Collapse
|
83
|
Magistri M, Velmeshev D, Makhmutova M, Faghihi MA. Transcriptomics Profiling of Alzheimer's Disease Reveal Neurovascular Defects, Altered Amyloid-β Homeostasis, and Deregulated Expression of Long Noncoding RNAs. J Alzheimers Dis 2016; 48:647-65. [PMID: 26402107 PMCID: PMC4698155 DOI: 10.3233/jad-150398] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The underlying genetic variations of late-onset Alzheimer's disease (LOAD) cases remain largely unknown. A combination of genetic variations with variable penetrance and lifetime epigenetic factors may converge on transcriptomic alterations that drive LOAD pathological process. Transcriptome profiling using deep sequencing technology offers insight into common altered pathways regardless of underpinning genetic or epigenetic factors and thus represents an ideal tool to investigate molecular mechanisms related to the pathophysiology of LOAD. We performed directional RNA sequencing on high quality RNA samples extracted from hippocampi of LOAD and age-matched controls. We further validated our data using qRT-PCR on a larger set of postmortem brain tissues, confirming downregulation of the gene encoding substance P (TAC1) and upregulation of the gene encoding the plasminogen activator inhibitor-1 (SERPINE1). Pathway analysis indicates dysregulation in neural communication, cerebral vasculature, and amyloid-β clearance. Beside protein coding genes, we identified several annotated and non-annotated long noncoding RNAs that are differentially expressed in LOAD brain tissues, three of them are activity-dependent regulated and one is induced by Aβ(1-42) exposure of human neural cells. Our data provide a comprehensive list of transcriptomics alterations in LOAD hippocampi and warrant holistic approach including both coding and non-coding RNAs in functional studies aimed to understand the pathophysiology of LOAD.
Collapse
Affiliation(s)
| | | | | | - Mohammad Ali Faghihi
- Correspondence to: Dr. Mohammad A. Faghihi, Center for Therapeutic Innovation & Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, 1501 NW 10th Ave, BRB 508, Miami, FL 33136, USA.
| |
Collapse
|
84
|
Toulorge D, Schapira AHV, Hajj R. Molecular changes in the postmortem parkinsonian brain. J Neurochem 2016; 139 Suppl 1:27-58. [PMID: 27381749 DOI: 10.1111/jnc.13696] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/14/2016] [Accepted: 05/27/2016] [Indexed: 12/16/2022]
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disease after Alzheimer disease. Although PD has a relatively narrow clinical phenotype, it has become clear that its etiological basis is broad. Post-mortem brain analysis, despite its limitations, has provided invaluable insights into relevant pathogenic pathways including mitochondrial dysfunction, oxidative stress and protein homeostasis dysregulation. Identification of the genetic causes of PD followed the discovery of these abnormalities, and reinforced the importance of the biochemical defects identified post-mortem. Recent genetic studies have highlighted the mitochondrial and lysosomal areas of cell function as particularly significant in mediating the neurodegeneration of PD. Thus the careful analysis of post-mortem PD brain biochemistry remains a crucial component of research, and one that offers considerable opportunity to pursue etiological factors either by 'reverse biochemistry' i.e. from defective pathway to mutant gene, or by the complex interplay between pathways e.g. mitochondrial turnover by lysosomes. In this review we have documented the spectrum of biochemical defects identified in PD post-mortem brain and explored their relevance to metabolic pathways involved in neurodegeneration. We have highlighted the complex interactions between these pathways and the gene mutations causing or increasing risk for PD. These pathways are becoming a focus for the development of disease modifying therapies for PD. Parkinson's is accompanied by multiple changes in the brain that are responsible for the progression of the disease. We describe here the molecular alterations occurring in postmortem brains and classify them as: Neurotransmitters and neurotrophic factors; Lewy bodies and Parkinson's-linked genes; Transition metals, calcium and calcium-binding proteins; Inflammation; Mitochondrial abnormalities and oxidative stress; Abnormal protein removal and degradation; Apoptosis and transduction pathways. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
| | | | - Rodolphe Hajj
- Department of Discovery, Pharnext, Issy-Les-Moulineaux, France.
| |
Collapse
|
85
|
Hervé M, Ibrahim EC. MicroRNA screening identifies a link between NOVA1 expression and a low level of IKAP in familial dysautonomia. Dis Model Mech 2016; 9:899-909. [PMID: 27483351 PMCID: PMC5007982 DOI: 10.1242/dmm.025841] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/22/2016] [Indexed: 12/20/2022] Open
Abstract
Familial dysautonomia (FD) is a rare neurodegenerative disease caused by a mutation in intron 20 of the IKBKAP gene (c.2204+6T>C), leading to tissue-specific skipping of exon 20 and a decrease in the synthesis of the encoded protein IKAP (also known as ELP1). Small non-coding RNAs known as microRNAs (miRNAs) are important post-transcriptional regulators of gene expression and play an essential role in the nervous system development and function. To better understand the neuronal specificity of IKAP loss, we examined expression of miRNAs in human olfactory ecto-mesenchymal stem cells (hOE-MSCs) from five control individuals and five FD patients. We profiled the expression of 373 miRNAs using microfluidics and reverse transcription coupled to quantitative PCR (RT-qPCR) on two biological replicate series of hOE-MSC cultures from healthy controls and FD patients. This led to the total identification of 26 dysregulated miRNAs in FD, validating the existence of a miRNA signature in FD. We then selected the nine most discriminant miRNAs for further analysis. The signaling pathways affected by these dysregulated miRNAs were largely within the nervous system. In addition, many targets of these dysregulated miRNAs had been previously demonstrated to be affected in FD models. Moreover, we found that four of our nine candidate miRNAs target the neuron-specific splicing factor NOVA1. We demonstrated that overexpression of miR-203a-3p leads to a decrease of NOVA1, counter-balanced by an increase of IKAP, supporting a potential interaction between NOVA1 and IKAP. Taken together, these results reinforce the choice of miRNAs as potential therapeutic targets and suggest that NOVA1 could be a regulator of FD pathophysiology. Summary: A miRNA screening conducted in olfactory stem cells from patients links the neuron-specific splicing factor NOVA1 to neurodegeneration in familial dysautonomia.
Collapse
Affiliation(s)
- Mylène Hervé
- CRN2M-UMR7286, Aix-Marseille Université, CNRS, Faculté de Médecine Nord, Marseille 13344, Cedex 15, France
| | - El Chérif Ibrahim
- CRN2M-UMR7286, Aix-Marseille Université, CNRS, Faculté de Médecine Nord, Marseille 13344, Cedex 15, France
| |
Collapse
|
86
|
Transcriptome profiling of the rat retina after optic nerve transection. Sci Rep 2016; 6:28736. [PMID: 27353354 PMCID: PMC4926057 DOI: 10.1038/srep28736] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023] Open
Abstract
Glaucoma is a group of eye diseases characterized by alterations in the contour of the optic nerve head (ONH), with corresponding visual field defects and progressive loss of retinal ganglion cells (RGCs). This progressive RGC death is considered to originate in axonal injury caused by compression of the axon bundles in the ONH. However, the molecular pathomechanisms of axonal injury-induced RGC death are not yet well understood. Here, we used RNA sequencing (RNA-seq) to examine transcriptome changes in rat retinas 2 days after optic nerve transection (ONT), and then used computational techniques to predict the resulting alterations in the transcriptional regulatory network. RNA-seq revealed 267 differentially expressed genes after ONT, 218 of which were annotated and 49 unannotated. We also identified differentially expressed transcripts, including potentially novel isoforms. An in silico pathway analysis predicted that CREB1 was the most significant upstream regulator. Thus, this study identified genes and pathways that may be involved in the pathomechanisms of axonal injury. We believe that our data should serve as a valuable resource to understand the molecular processes that define axonal injury-driven RGC death and to discover novel therapeutic targets for glaucoma.
Collapse
|
87
|
Chen X, Petranovic D. Role of frameshift ubiquitin B protein in Alzheimer's disease. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2016; 8:300-13. [DOI: 10.1002/wsbm.1340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/10/2016] [Accepted: 03/19/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Xin Chen
- Systems and Synthetic Biology, Department of Biology and Biological Engineering; Chalmers University of Technology; Göteborg Sweden
| | - Dina Petranovic
- Systems and Synthetic Biology, Department of Biology and Biological Engineering; Chalmers University of Technology; Göteborg Sweden
- Novo Nordisk Foundation Center for Biosustainability; Chalmers University of Technology; Göteborg Sweden
| |
Collapse
|
88
|
Henderson-Smith A, Corneveaux JJ, De Both M, Cuyugan L, Liang WS, Huentelman M, Adler C, Driver-Dunckley E, Beach TG, Dunckley TL. Next-generation profiling to identify the molecular etiology of Parkinson dementia. NEUROLOGY-GENETICS 2016; 2:e75. [PMID: 27275011 PMCID: PMC4881621 DOI: 10.1212/nxg.0000000000000075] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 03/21/2016] [Indexed: 12/15/2022]
Abstract
OBJECTIVE We sought to determine the underlying cortical gene expression changes associated with Parkinson dementia using a next-generation RNA sequencing approach. METHODS In this study, we used RNA sequencing to evaluate differential gene expression and alternative splicing in the posterior cingulate cortex from neurologically normal control patients, patients with Parkinson disease, and patients with Parkinson disease with dementia. RESULTS Genes overexpressed in both disease states were involved with an immune response, whereas shared underexpressed genes functioned in signal transduction or as components of the cytoskeleton. Alternative splicing analysis produced a pattern of immune and RNA-processing disturbances. CONCLUSIONS Genes with the greatest degree of differential expression did not overlap with genes exhibiting significant alternative splicing activity. Such variation indicates the importance of broadening expression studies to include exon-level changes because there can be significant differential splicing activity with potential structural consequences, a subtlety that is not detected when examining differential gene expression alone, or is underrepresented with probe-limited array technology.
Collapse
Affiliation(s)
- Adrienne Henderson-Smith
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Jason J Corneveaux
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Matthew De Both
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Lori Cuyugan
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Winnie S Liang
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Matthew Huentelman
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Charles Adler
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Erika Driver-Dunckley
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Thomas G Beach
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| | - Travis L Dunckley
- Neurogenomics Division (A.H.-S., J.J.C., M.D.B., L.C., W.S.L., M.H., T.L.D.), Collaborative Sequencing Center (L.C., W.S.L.), Translational Genomics Research Institute, Phoenix; Division of Neurology (C.A., E.D.-D.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B.), Sun City, AZ
| |
Collapse
|
89
|
Ciryam P, Kundra R, Freer R, Morimoto RI, Dobson CM, Vendruscolo M. A transcriptional signature of Alzheimer's disease is associated with a metastable subproteome at risk for aggregation. Proc Natl Acad Sci U S A 2016; 113:4753-8. [PMID: 27071083 PMCID: PMC4855616 DOI: 10.1073/pnas.1516604113] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
It is well-established that widespread transcriptional changes accompany the onset and progression of Alzheimer's disease. Because of the multifactorial nature of this neurodegenerative disorder and its complex relationship with aging, however, it remains unclear whether such changes are the result of nonspecific dysregulation and multisystem failure or instead are part of a coordinated response to cellular dysfunction. To address this problem in a systematic manner, we performed a meta-analysis of about 1,600 microarrays from human central nervous system tissues to identify transcriptional changes upon aging and as a result of Alzheimer's disease. Our strategy to discover a transcriptional signature of Alzheimer's disease revealed a set of down-regulated genes that encode proteins metastable to aggregation. Using this approach, we identified a small number of biochemical pathways, notably oxidative phosphorylation, enriched in proteins vulnerable to aggregation in control brains and encoded by genes down-regulated in Alzheimer's disease. These results suggest that the down-regulation of a metastable subproteome may help mitigate aberrant protein aggregation when protein homeostasis becomes compromised in Alzheimer's disease.
Collapse
Affiliation(s)
- Prajwal Ciryam
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom; Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Rishika Kundra
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Rosie Freer
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Richard I Morimoto
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Christopher M Dobson
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Michele Vendruscolo
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom;
| |
Collapse
|
90
|
Wagner A, Cohen N, Kelder T, Amit U, Liebman E, Steinberg DM, Radonjic M, Ruppin E. Drugs that reverse disease transcriptomic signatures are more effective in a mouse model of dyslipidemia. Mol Syst Biol 2016; 11:791. [PMID: 26148350 PMCID: PMC4380926 DOI: 10.15252/msb.20145486] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
High-throughput omics have proven invaluable in studying human disease, and yet day-to-day clinical practice still relies on physiological, non-omic markers. The metabolic syndrome, for example, is diagnosed and monitored by blood and urine indices such as blood cholesterol levels. Nevertheless, the association between the molecular and the physiological manifestations of the disease, especially in response to treatment, has not been investigated in a systematic manner. To this end, we studied a mouse model of diet-induced dyslipidemia and atherosclerosis that was subject to various drug treatments relevant to the disease in question. Both physiological data and gene expression data (from the liver and white adipose) were analyzed and compared. We find that treatments that restore gene expression patterns to their norm are associated with the successful restoration of physiological markers to their baselines. This holds in a tissue-specific manner—treatments that reverse the transcriptomic signatures of the disease in a particular tissue are associated with positive physiological effects in that tissue. Further, treatments that introduce large non-restorative gene expression alterations are associated with unfavorable physiological outcomes. These results provide a sound basis to in silico methods that rely on omic metrics for drug repurposing and drug discovery by searching for compounds that reverse a disease's omic signatures. Moreover, they highlight the need to develop drugs that restore the global cellular state to its healthy norm rather than rectify particular disease phenotypes.
Collapse
Affiliation(s)
- Allon Wagner
- The Blavatnik School of Computer Science, Tel Aviv UniversityTel Aviv, Israel
- Department of Electrical Engineering and Computer Science, University of CaliforniaBerkeley, CA, USA
- * Corresponding author. Tel. +972 3 640 5378; E-mail:
| | - Noa Cohen
- The Blavatnik School of Computer Science, Tel Aviv UniversityTel Aviv, Israel
| | - Thomas Kelder
- Microbiology and Systems Biology, TNOZeist, the Netherlands
| | - Uri Amit
- Neufeld Cardiac Research Institute, Tel Aviv UniversityTel Aviv, Israel
- Regenerative Medicine Stem Cells and Tissue Engineering Center, Sheba Medical CenterTel Hashomer, Israel
| | - Elad Liebman
- Department of Computer Science, University of Texas at AustinAustin, TX, USA
| | - David M Steinberg
- Department of Statistics and Operations Research, Tel Aviv UniversityTel Aviv, Israel
| | | | - Eytan Ruppin
- The Blavatnik School of Computer Science, Tel Aviv UniversityTel Aviv, Israel
- The Sackler School of Medicine, Tel Aviv UniversityTel Aviv, Israel
- Department of Computer Science, Institute of Advanced Computer Sciences (UMIACS) & the Center for Bioinformatics and Computational Biology, University of MarylandCollege Park, MD, USA
- ** Corresponding author. Tel. +972 3 640 6528; E-mail:
| |
Collapse
|
91
|
Gene dysregulation is restored in the Parkinson's disease MPTP neurotoxic mice model upon treatment of the therapeutic drug Cu(II)(atsm). Sci Rep 2016; 6:22398. [PMID: 26928495 PMCID: PMC4772163 DOI: 10.1038/srep22398] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 02/15/2016] [Indexed: 12/04/2022] Open
Abstract
The administration of MPTP selectively targets the dopaminergic system resulting in Parkinsonism-like symptoms and is commonly used as a mice model of Parkinson’s disease. We previously demonstrated that the neuroprotective compound CuII(atsm) rescues nigral cell loss and improves dopamine metabolism in the MPTP model. The mechanism of action of CuII(atsm) needs to be further defined to understand how the compound promotes neuronal survival. Whole genome transcriptomic profiling has become a popular method to examine the relationship between gene expression and function. Substantia nigra samples from MPTP-lesioned mice were evaluated using whole transcriptome sequencing to investigate the genes altered upon CuII(atsm) treatment. We identified 143 genes affected by MPTP lesioning that are associated with biological processes related to brain and cognitive development, dopamine synthesis and perturbed synaptic neurotransmission. Upon CuII(atsm) treatment, the expression of 40 genes involved in promoting dopamine synthesis, calcium signaling and synaptic plasticity were restored which were validated by qRT-PCR. The study provides the first detailed whole transcriptomic analysis of pathways involved in MPTP-induced Parkinsonism. In addition, we identify key therapeutic pathways targeted by a potentially new class of neuroprotective agents which may provide therapeutic benefits for other neurodegenerative disorders.
Collapse
|
92
|
Investigating the impact human protein–protein interaction networks have on disease-gene analysis. INT J MACH LEARN CYB 2016. [DOI: 10.1007/s13042-016-0503-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
93
|
Deep brain stimulation-associated brain tissue imprints: a new in vivo approach to biological research in human Parkinson's disease. Mol Neurodegener 2016; 11:12. [PMID: 26822202 PMCID: PMC4730746 DOI: 10.1186/s13024-016-0077-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/20/2016] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Deep brain stimulation (DBS) of the subthalamic nucleus (STN) or the internal segment of the globus pallidus (GPi) has been established as a highly effective symptomatic therapy for Parkinson's disease (PD). An intriguing biological aspect related to the DBS procedure is that a temporary contact establishes between surgical instruments and the surrounding brain tissue. In this exploratory study, we took advantage of this unique context to harvest brain material adhering to the stylet routinely used during surgery, and to examine the biological value of these samples, here referred to as "brain tissue imprints" (BTIs). RESULTS Nineteen BTIs from 12 STN- or GPi-electrode implanted patients were obtained in vivo during DBS surgery, without any modification of the surgical procedure. Immunofluorescence analyses confirmed that our approach allowed the harvesting of many neural cells including neurons harboring distinct neurotransmitter markers. Shotgun proteomic and transcriptomic analyses provided for the first time molecular information from DBS-associated brain samples, and confirmed the compatibility of this new type of sample with poly-omic approaches. The method appears to be safe and results consistent. CONCLUSIONS We here propose BTIs as original and highly valuable brain samples, and DBS-related brain imprinting as a new conceptual approach to biological research in living patients with PD.
Collapse
|
94
|
Oh SM, Chang MY, Song JJ, Rhee YH, Joe EH, Lee HS, Yi SH, Lee SH. Combined Nurr1 and Foxa2 roles in the therapy of Parkinson's disease. EMBO Mol Med 2016; 7:510-25. [PMID: 25759364 PMCID: PMC4492814 DOI: 10.15252/emmm.201404610] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Use of the physiological mechanisms promoting midbrain DA (mDA) neuron survival seems an appropriate option for developing treatments for Parkinson's disease (PD). mDA neurons are specifically marked by expression of the transcription factors Nurr1 and Foxa2. We show herein that Nurr1 and Foxa2 interact to protect mDA neurons against various toxic insults, but their expression is lost during aging and degenerative processes. In addition to their proposed cell-autonomous actions in mDA neurons, forced expression of these factors in neighboring glia synergistically protects degenerating mDA neurons in a paracrine mode. As a consequence of these bimodal actions, adeno-associated virus (AAV)-mediated gene delivery of Nurr1 and Foxa2 in a PD mouse model markedly protected mDA neurons and motor behaviors associated with nigrostriatal DA neurotransmission. The effects of the combined gene delivery were dramatic, highly reproducible, and sustained for at least 1 year, suggesting that expression of these factors is a promising approach in PD therapy.
Collapse
Affiliation(s)
- Sang-Min Oh
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Korea Hanyang Biomedical Research Institute, Hanyang University, Seoul, Korea Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Mi-Yoon Chang
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Jae-Jin Song
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Korea Hanyang Biomedical Research Institute, Hanyang University, Seoul, Korea Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Yong-Hee Rhee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Korea Hanyang Biomedical Research Institute, Hanyang University, Seoul, Korea Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Eun-Hye Joe
- Department of Phamacology, Ajou University School of Medicine, Suwon, Korea
| | - Hyun-Seob Lee
- Department of Applied Bioscience, College of Life Science, CHA University, Seoul, Korea
| | - Sang-Hoon Yi
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Sang-Hun Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Korea Hanyang Biomedical Research Institute, Hanyang University, Seoul, Korea Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| |
Collapse
|
95
|
MacNair L, Xiao S, Miletic D, Ghani M, Julien JP, Keith J, Zinman L, Rogaeva E, Robertson J. MTHFSD and DDX58 are novel RNA-binding proteins abnormally regulated in amyotrophic lateral sclerosis. Brain 2015; 139:86-100. [PMID: 26525917 DOI: 10.1093/brain/awv308] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/03/2015] [Indexed: 01/21/2023] Open
Abstract
Tar DNA-binding protein 43 (TDP-43) is an RNA-binding protein normally localized to the nucleus of cells, where it elicits functions related to RNA metabolism such as transcriptional regulation and alternative splicing. In amyotrophic lateral sclerosis, TDP-43 is mislocalized from the nucleus to the cytoplasm of diseased motor neurons, forming ubiquitinated inclusions. Although mutations in the gene encoding TDP-43, TARDBP, are found in amyotrophic lateral sclerosis, these are rare. However, TDP-43 pathology is common to over 95% of amyotrophic lateral sclerosis cases, suggesting that abnormalities of TDP-43 play an active role in disease pathogenesis. It is our hypothesis that a loss of TDP-43 from the nucleus of affected motor neurons in amyotrophic lateral sclerosis will lead to changes in RNA processing and expression. Identifying these changes could uncover molecular pathways that underpin motor neuron degeneration. Here we have used translating ribosome affinity purification coupled with microarray analysis to identify the mRNAs being actively translated in motor neurons of mutant TDP-43(A315T) mice compared to age-matched non-transgenic littermates. No significant changes were found at 5 months (presymptomatic) of age, but at 10 months (symptomatic) the translational profile revealed significant changes in genes involved in RNA metabolic process, immune response and cell cycle regulation. Of 28 differentially expressed genes, seven had a ≥ 2-fold change; four were validated by immunofluorescence labelling of motor neurons in TDP-43(A315T) mice, and two of these were confirmed by immunohistochemistry in amyotrophic lateral sclerosis cases. Both of these identified genes, DDX58 and MTHFSD, are RNA-binding proteins, and we show that TDP-43 binds to their respective mRNAs and we identify MTHFSD as a novel component of stress granules. This discovery-based approach has for the first time revealed translational changes in motor neurons of a TDP-43 mouse model, identifying DDX58 and MTHFSD as two TDP-43 targets that are misregulated in amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Laura MacNair
- 1 Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 2S8, Canada 2 Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Shangxi Xiao
- 1 Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 2S8, Canada
| | - Denise Miletic
- 1 Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 2S8, Canada
| | - Mahdi Ghani
- 1 Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 2S8, Canada
| | - Jean-Pierre Julien
- 3 Département de psychiatrie et de neurosciences, Université Laval, Québec G1V 0A6, Canada
| | - Julia Keith
- 2 Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A1, Canada 4 Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Lorne Zinman
- 4 Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | - Ekaterina Rogaeva
- 1 Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 2S8, Canada
| | - Janice Robertson
- 1 Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 2S8, Canada 2 Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A1, Canada
| |
Collapse
|
96
|
Comparative blood transcriptome analysis in idiopathic and LRRK2 G2019S-associated Parkinson's disease. Neurobiol Aging 2015; 38:214.e1-214.e5. [PMID: 26675812 DOI: 10.1016/j.neurobiolaging.2015.10.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 09/16/2015] [Accepted: 10/27/2015] [Indexed: 12/21/2022]
Abstract
Patients with Parkinson's disease (PD) carrying the G2019S mutation of the LRRK2 gene provide an opportunity of studying in a homogeneous setting the molecular pathways involved in the pathogenesis of common idiopathic forms of PD. However, whether common mechanisms are involved in both conditions in not known. Here, we compared genome-wide gene expression (RNA sequencing) in peripheral blood between PD patients carrying the G2019S mutation of the LRRK2 gene and idiopathic PD cases, to deepen in the understanding of this topic. In addition, we compared the blood transcriptome between 2 cohorts of carriers of the G2019S mutation (symptomatic and asymptomatic) and 2 cohorts of noncarriers (symptomatic and asymptomatic) for detecting transcriptomic changes attributable to the presence of the G2019S mutation. We searched for gene enrichment in Reactome or Kyoto Encyclopedia of Genes and Genomes pathways. We found that despite some overlap, peripheral blood transcriptome differs widely between idiopathic and LRRK2 G2019S-associated PD, with only 4 deregulated pathways shared by both conditions (complement and coagulation cascades, cell adhesion molecules, hematopoietic cell lineage, and extracellular matrix organization). Changes in the blood transcriptome observed in asymptomatic carriers of the mutation included 6 genes known to be associated with PD in genome-wide association studies and also pathways related with immunity. Our findings emphasize the notion that PD is likely a pathogenically heterogeneous condition and suggest the existence of specific mechanisms involved in LRRK2-associated PD.
Collapse
|
97
|
Haston KM, Finkbeiner S. Clinical Trials in a Dish: The Potential of Pluripotent Stem Cells to Develop Therapies for Neurodegenerative Diseases. Annu Rev Pharmacol Toxicol 2015; 56:489-510. [PMID: 26514199 PMCID: PMC4868344 DOI: 10.1146/annurev-pharmtox-010715-103548] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases are a leading cause of death. No disease-modifying therapies are available, and preclinical animal model data have routinely failed to translate into success for therapeutics. Induced pluripotent stem cell (iPSC) biology holds great promise for human in vitro disease modeling because these cells can give rise to any cell in the human brain and display phenotypes specific to neurodegenerative diseases previously identified in postmortem and clinical samples. Here, we explore the potential and caveats of iPSC technology as a platform for drug development and screening, and the future potential to use large cohorts of disease-bearing iPSCs to perform clinical trials in a dish.
Collapse
Affiliation(s)
- Kelly M Haston
- Gladstone Institute of Neurological Disease, San Francisco, California 94158;
| | - Steven Finkbeiner
- Gladstone Institute of Neurological Disease, San Francisco, California 94158;
- Taube/Koret Center for Neurodegenerative Disease and the Hellman Family Foundation Program in Alzheimer's Disease Research, San Francisco, California 94158
- Departments of Neurology and Physiology, University of California, San Francisco, California 94143
| |
Collapse
|
98
|
Violet M, Chauderlier A, Delattre L, Tardivel M, Chouala MS, Sultan A, Marciniak E, Humez S, Binder L, Kayed R, Lefebvre B, Bonnefoy E, Buée L, Galas MC. Prefibrillar Tau oligomers alter the nucleic acid protective function of Tau in hippocampal neurons in vivo. Neurobiol Dis 2015; 82:540-551. [PMID: 26385829 DOI: 10.1016/j.nbd.2015.09.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 07/09/2015] [Accepted: 09/13/2015] [Indexed: 01/05/2023] Open
Abstract
The accumulation of DNA and RNA oxidative damage is observed in cortical and hippocampal neurons from Alzheimer's disease (AD) brains at early stages of pathology. We recently reported that Tau is a key nuclear player in the protection of neuronal nucleic acid integrity in vivo under physiological conditions and hyperthermia, a strong inducer of oxidative stress. In a mouse model of tauopathy (THY-Tau22), we demonstrate that hyperthermia selectively induces nucleic acid oxidative damage and nucleic acid strand breaks in the nucleus and cytoplasm of hippocampal neurons that display early Tau phosphorylation but no Tau fibrils. Nucleic acid-damaged neurons were exclusively immunoreactive for prefibrillar Tau oligomers. A similar association between prefibrillar Tau oligomers and nucleic acid oxidative damage was observed in AD brains. Pretreatment with Methylene Blue (MB), a Tau aggregation inhibitor and a redox cycler, reduced hyperthermia-induced Tau oligomerization as well as nucleic acid damage. This study clearly highlights the existence of an early and critical time frame for hyperthermia-induced Tau oligomerization, which most likely occurs through increased oxidative stress, and nucleic acid vulnerability during the progression of Tau pathology. These results suggest that at early stages of AD, Tau oligomerization triggers the loss of the nucleic acid protective function of monomeric Tau. This study highlights the existence of a short therapeutic window in which to prevent the formation of pathological forms of Tau and their harmful consequences on nucleic acid integrity during the progression of Tau pathology.
Collapse
Affiliation(s)
- Marie Violet
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Alban Chauderlier
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Lucie Delattre
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Meryem Tardivel
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Meliza Sendid Chouala
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Audrey Sultan
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Elodie Marciniak
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Sandrine Humez
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Lester Binder
- Department of Translational Science & Molecular Medicine, College of Human Medicine, Michigan State University, 333 Bostwick Ave. NE, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Rakez Kayed
- Department of Neurology, George and Cynthia Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Medical Research Building, Room 10.138C, Galveston, TX 77555-1045, USA; Department of Neuroscience & Cell Biology, George and Cynthia Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Medical Research Building, Room 10.138C, Galveston, TX 77555-1045, USA
| | - Bruno Lefebvre
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Eliette Bonnefoy
- Inserm UMRS 1007, Université Paris Descartes, 45 rue des Saints Pères, 75006 Paris Cedex 06, France
| | - Luc Buée
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Marie-Christine Galas
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France.
| |
Collapse
|
99
|
Wang L, Wu LF, Lu X, Mo XB, Tang ZX, Lei SF, Deng FY. Integrated Analyses of Gene Expression Profiles Digs out Common Markers for Rheumatic Diseases. PLoS One 2015; 10:e0137522. [PMID: 26352601 PMCID: PMC4564267 DOI: 10.1371/journal.pone.0137522] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 08/18/2015] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE Rheumatic diseases have some common symptoms. Extensive gene expression studies, accumulated thus far, have successfully identified signature molecules for each rheumatic disease, individually. However, whether there exist shared factors across rheumatic diseases has yet to be tested. METHODS We collected and utilized 6 public microarray datasets covering 4 types of representative rheumatic diseases including rheumatoid arthritis, systemic lupus erythematosus, ankylosing spondylitis, and osteoarthritis. Then we detected overlaps of differentially expressed genes across datasets and performed a meta-analysis aiming at identifying common differentially expressed genes that discriminate between pathological cases and normal controls. To further gain insights into the functions of the identified common differentially expressed genes, we conducted gene ontology enrichment analysis and protein-protein interaction analysis. RESULTS We identified a total of eight differentially expressed genes (TNFSF10, CX3CR1, LY96, TLR5, TXN, TIA1, PRKCH, PRF1), each associated with at least 3 of the 4 studied rheumatic diseases. Meta-analysis warranted the significance of the eight genes and highlighted the general significance of four genes (CX3CR1, LY96, TLR5, and PRF1). Protein-protein interaction and gene ontology enrichment analyses indicated that the eight genes interact with each other to exert functions related to immune response and immune regulation. CONCLUSION The findings support that there exist common factors underlying rheumatic diseases. For rheumatoid arthritis, systemic lupus erythematosus, ankylosing spondylitis and osteoarthritis diseases, those common factors include TNFSF10, CX3CR1, LY96, TLR5, TXN, TIA1, PRKCH, and PRF1. In-depth studies on these common factors may provide keys to understanding the pathogenesis and developing intervention strategies for rheumatic diseases.
Collapse
Affiliation(s)
- Lan Wang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Long-Fei Wu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Xin Lu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Xing-Bo Mo
- Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Zai-Xiang Tang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Shu-Feng Lei
- Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Fei-Yan Deng
- Center for Genetic Epidemiology and Genomics, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, Jiangsu 215123, P. R. China
- * E-mail:
| |
Collapse
|
100
|
Comparative Incidence of Conformational, Neurodegenerative Disorders. PLoS One 2015; 10:e0137342. [PMID: 26335347 PMCID: PMC4559310 DOI: 10.1371/journal.pone.0137342] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/14/2015] [Indexed: 12/12/2022] Open
Abstract
Background The purpose of this study was to identify incidence and survival patterns in conformational neurodegenerative disorders (CNDDs). Methods We identified 2563 reports on the incidence of eight conditions representing sporadic, acquired and genetic, protein-associated, i.e., conformational, NDD groups and age-related macular degeneration (AMD). We selected 245 papers for full-text examination and application of quality criteria. Additionally, data-collection was completed with detailed information from British, Swedish, and Spanish registries on Creutzfeldt-Jakob disease (CJD) forms, amyotrophic lateral sclerosis (ALS), and sporadic rapidly progressing neurodegenerative dementia (sRPNDd). For each condition, age-specific incidence curves, age-adjusted figures, and reported or calculated median survival were plotted and examined. Findings Based on 51 valid reported and seven new incidence data sets, nine out of eleven conditions shared specific features. Age-adjusted incidence per million person-years increased from ≤1.5 for sRPNDd, different CJD forms and Huntington's disease (HD), to 1589 and 2589 for AMD and Alzheimer's disease (AD) respectively. Age-specific profiles varied from (a) symmetrical, inverted V-shaped curves for low incidences to (b) those increasing with age for late-life sporadic CNDDs and for sRPNDd, with (c) a suggested, intermediate, non-symmetrical inverted V-shape for fronto-temporal dementia and Parkinson's disease. Frequently, peak age-specific incidences from 20–24 to ≥90 years increased with age at onset and survival. Distinct patterns were seen: for HD, with a low incidence, levelling off at middle age, and long median survival, 20 years; and for sRPNDd which displayed the lowest incidence, increasing with age, and a short median disease duration. Interpretation These results call for a unified population view of NDDs, with an age-at-onset-related pattern for acquired and sporadic CNDDs. The pattern linking age at onset to incidence magnitude and survival might be explained by differential pathophysiological mechanisms associated with specific misfolded protein deposits.
Collapse
|