51
|
Wachinger C, Nho K, Saykin AJ, Reuter M, Rieckmann A. A Longitudinal Imaging Genetics Study of Neuroanatomical Asymmetry in Alzheimer's Disease. Biol Psychiatry 2018; 84:522-530. [PMID: 29885764 PMCID: PMC6123250 DOI: 10.1016/j.biopsych.2018.04.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/25/2018] [Accepted: 04/25/2018] [Indexed: 11/18/2022]
Abstract
BACKGROUND Contralateral brain structures represent a unique, within-patient reference element for disease, and asymmetries can provide a personalized measure of the accumulation of past disease processes. Neuroanatomical shape asymmetries have recently been associated with the progression of Alzheimer's disease (AD), but the biological basis of asymmetric brain changes in AD remains unknown. METHODS We investigated genetic influences on brain asymmetry by identifying associations between magnetic resonance imaging-derived measures of asymmetry and candidate single nucleotide polymorphisms (SNPs) that have previously been identified in genome-wide association studies for AD diagnosis and for brain subcortical volumes. For analyzing longitudinal neuroimaging data (1241 individuals, 6395 scans), we used a mixed effects model with interaction between genotype and diagnosis. RESULTS Significant associations between asymmetry of the amygdala, hippocampus, and putamen and SNPs in the genes BIN1, CD2AP, ZCWPW1, ABCA7, TNKS, and DLG2 were found. CONCLUSIONS The associations between SNPs in the genes TNKS and DLG2 and AD-related increases in shape asymmetry are of particular interest; these SNPs have previously been associated with subcortical volumes of amygdala and putamen but have not yet been associated with AD pathology. For AD candidate SNPs, we extend previous work to show that their effects on subcortical brain structures are asymmetric. This provides novel evidence about the biological underpinnings of brain asymmetry as a disease marker.
Collapse
Affiliation(s)
- Christian Wachinger
- Laboratory for Artificial Intelligence in Medical Imaging, Klinik für Kinder- und Jugendpsychiatrie, Klinikum der Universität München, Ludwig-Maximilians-Universität München, München, Germany.
| | - Kwangsik Nho
- Center for Neuroimaging and Indiana Alzheimer Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| | - Andrew J Saykin
- Center for Neuroimaging and Indiana Alzheimer Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| | - Martin Reuter
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts; Deutsches Zentrum für Neurodegenerative Erkrankungen, Bonn, Germany
| | - Anna Rieckmann
- Umeå Center for Functional Brain Imaging, Department of Radiation Sciences, Umeå University, Umeå, Sweden
| |
Collapse
|
52
|
Inherited and Acquired Decrease in Complement Receptor 1 (CR1) Density on Red Blood Cells Associated with High Levels of Soluble CR1 in Alzheimer's Disease. Int J Mol Sci 2018; 19:ijms19082175. [PMID: 30044434 PMCID: PMC6121509 DOI: 10.3390/ijms19082175] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/22/2018] [Accepted: 07/23/2018] [Indexed: 12/13/2022] Open
Abstract
The complement receptor 1 (CR1) gene was shown to be involved in Alzheimer's disease (AD). We previously showed that AD is associated with low density of the long CR1 isoform, CR1*2 (S). Here, we correlated phenotype data (CR1 density per erythrocyte (CR1/E), blood soluble CR1 (sCR1)) with genetic data (density/length polymorphisms) in AD patients and healthy controls. CR1/E was enumerated using flow cytometry, while sCR1 was quantified by ELISA. CR1 polymorphisms were assessed using restriction fragment length polymorphism (RFLP), pyrosequencing, and high-resolution melting PCR. In AD patients carrying the H allele (HindIII polymorphism) or the Q allele (Q981H polymorphism), CR1/E was significantly lower when compared with controls carrying the same alleles (p < 0.01), contrary to sCR1, which was significantly higher (p < 0.001). Using multivariate analysis, a reduction of 6.68 units in density was associated with an increase of 1% in methylation of CR1 (estimate -6.68; 95% confidence intervals (CIs) -12.37, -0.99; p = 0.02). Our data show that, in addition to inherited genetic factors, low density of CR1/E is also acquired. The involvement of CR1 in the pathogenesis of AD might be linked to insufficient clearance of amyloid deposits. These findings may open perspectives for new therapeutic strategies in AD.
Collapse
|
53
|
Huang ZH, Chen HC, Chou YC, Lin CL, Kao CH, Lo HY, Yang TY, Liu FC. Does nephrotic syndrome without chronic kidney disease increase the risk of Parkinson's disease and secondary parkinsonism? A nationwide population-based study in Taiwan. BMJ Open 2018; 8:e020821. [PMID: 29982207 PMCID: PMC6045768 DOI: 10.1136/bmjopen-2017-020821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 04/27/2018] [Accepted: 06/08/2018] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVES Previous research has shown that patients with nephrotic syndrome (NS) have a higher risk of cognitive impairment, dementia or neurodegenerative disorder. The present study aimed to examine a relationship, if any exists between NS and Parkinson's disease (PD), a neurodegenerative disorder and secondary parkinsonism (sPS). METHODS A nationwide retrospective observational study conducted using data from the 2000-2010 Taiwan National Health Insurance Research Database. This study included 3663 patients with NS and 14 652 randomly selected, age-matched and sex-matched patients without NS. A Cox multivariable proportional hazards model was used to evaluate the risk of PD and sPS (PDsPS) in the NS cohort. RESULTS This study identified a positive association between NS and the risk of PDsPS in both men and women and in all age groups (adjusted HR 1.51; 95% CI 1.37 to 1.66). Compared with patients without NS and comorbidities, those with NS with two or more comorbidities exhibited an 8.23-fold higher risk of PDsPS (95% CI 6.22 to 10.9) and patients with NS and one comorbidity exhibited a 2.93-fold higher risk of PDsPS (95% CI 2.37 to 3.63). CONCLUSIONS Patients with NS have an increased risk of PDsPS. This increased risk may be related to brain vascular damage or blood-brain barrier impairment. Further research is necessary to explore the underlying relationship between NS and PDsPS.
Collapse
Affiliation(s)
- Zheng-Hao Huang
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Hsiang-Cheng Chen
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Ching Chou
- Department of Health Promotion and Health Education, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Li Lin
- School of Medicine, China Medical University, Taichung, Taiwan
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Hung Kao
- Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Clinical Medicine Science and School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Hsin-Yi Lo
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Tse-Yen Yang
- Molecular and Genomic Epidemiology Center, China Medical University Hospital, Taichung, Taiwan
| | - Feng-Cheng Liu
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
54
|
Jeong A, Suazo KF, Wood WG, Distefano MD, Li L. Isoprenoids and protein prenylation: implications in the pathogenesis and therapeutic intervention of Alzheimer's disease. Crit Rev Biochem Mol Biol 2018; 53:279-310. [PMID: 29718780 PMCID: PMC6101676 DOI: 10.1080/10409238.2018.1458070] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The mevalonate-isoprenoid-cholesterol biosynthesis pathway plays a key role in human health and disease. The importance of this pathway is underscored by the discovery that two major isoprenoids, farnesyl and geranylgeranyl pyrophosphate, are required to modify an array of proteins through a process known as protein prenylation, catalyzed by prenyltransferases. The lipophilic prenyl group facilitates the anchoring of proteins in cell membranes, mediating protein-protein interactions and signal transduction. Numerous essential intracellular proteins undergo prenylation, including most members of the small GTPase superfamily as well as heterotrimeric G proteins and nuclear lamins, and are involved in regulating a plethora of cellular processes and functions. Dysregulation of isoprenoids and protein prenylation is implicated in various disorders, including cardiovascular and cerebrovascular diseases, cancers, bone diseases, infectious diseases, progeria, and neurodegenerative diseases including Alzheimer's disease (AD). Therefore, isoprenoids and/or prenyltransferases have emerged as attractive targets for developing therapeutic agents. Here, we provide a general overview of isoprenoid synthesis, the process of protein prenylation and the complexity of prenylated proteins, and pharmacological agents that regulate isoprenoids and protein prenylation. Recent findings that connect isoprenoids/protein prenylation with AD are summarized and potential applications of new prenylomic technologies for uncovering the role of prenylated proteins in the pathogenesis of AD are discussed.
Collapse
Affiliation(s)
- Angela Jeong
- Departments of Experimental and Clinical Pharmacolog,University of Minnesota, Minneapolis, MN 55455
| | | | - W. Gibson Wood
- Departments of Pharmacology, University of Minnesota, Minneapolis, MN 55455
| | - Mark D. Distefano
- Departments of Chemistry,University of Minnesota, Minneapolis, MN 55455
| | - Ling Li
- Departments of Experimental and Clinical Pharmacolog,University of Minnesota, Minneapolis, MN 55455
- Departments of Pharmacology, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
55
|
Michno W, Kaya I, Nyström S, Guerard L, Nilsson KPR, Hammarström P, Blennow K, Zetterberg H, Hanrieder J. Multimodal Chemical Imaging of Amyloid Plaque Polymorphism Reveals Aβ Aggregation Dependent Anionic Lipid Accumulations and Metabolism. Anal Chem 2018; 90:8130-8138. [DOI: 10.1021/acs.analchem.8b01361] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Wojciech Michno
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Ibrahim Kaya
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Sofie Nyström
- IFM-Department of Chemistry, Linköping University, Linköping, Sweden
| | - Laurent Guerard
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- IMCF Biozentrum, University of Basel, Basel, Switzerland
| | | | - Per Hammarström
- IFM-Department of Chemistry, Linköping University, Linköping, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
56
|
Huovinen J, Helisalmi S, Paananen J, Laiterä T, Kojoukhova M, Sutela A, Vanninen R, Laitinen M, Rauramaa T, Koivisto AM, Remes AM, Soininen H, Kurki M, Haapasalo A, Jääskeläinen JE, Hiltunen M, Leinonen V. Alzheimer's Disease-Related Polymorphisms in Shunt-Responsive Idiopathic Normal Pressure Hydrocephalus. J Alzheimers Dis 2018; 60:1077-1085. [PMID: 28984604 DOI: 10.3233/jad-170583] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Idiopathic normal pressure hydrocephalus (iNPH) is a late onset, surgically treated progressive brain disease caused by impaired cerebrospinal fluid dynamics and subsequent ventriculomegaly. Comorbid Alzheimer's disease (AD) seems to be frequent in iNPH. OBJECTIVE We aim to evaluate the role of AD-related polymorphisms in iNPH. METHODS Overall 188 shunt-operated iNPH patients and 688 controls without diagnosed neurodegenerative disease were included into analysis. Twenty-three single-nucleotide polymorphisms (SNPs FRMD4A [rs7081208_A, rs2446581_A, rs17314229_T], CR1, BIN, CD2AP, CLU, MS4A6A, MS4A4E, PICALM, ABCA7, CD33, INPP5D, HLA_DRB5, EPHA1, PTK2B, CELF1, SORL1, FERMT2, SLC24A, DSG2, CASS4, and NME8) adjusted to APOE were analyzed between groups by using binary logistic regression analysis. Neuroradiological characteristics and AD-related changes in the right frontal cortical brain biopsies were available for further analysis. RESULTS Logistic regression analysis adjusted to age, gender, and other SNPs indicated allelic variation of NME8 between iNPH patients and non-demented controls (p = 0.014). The allelic variation of NME8 was not related to the neuropathological changes in the brain biopsies of iNPH patients. However, periventricular white matter changes (p = 0.017) were more frequent in the iNPH patients with the AA-genotype, an identified risk factor of AD. CONCLUSIONS Our findings increase the evidence that iNPH is characterized by genetic and pathophysiological mechanisms independent from AD. Considering that NME8 plays a role in the ciliary function and displays SNP-related diversity in white matter changes, the mechanisms of NME8 in iNPH and other neurodegenerative processes are worth further study.
Collapse
Affiliation(s)
- Joel Huovinen
- Institute of Clinical Medicine -Neurosurgery, University of Eastern Finland and Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland
| | - Seppo Helisalmi
- Institute of Clinical Medicine -Neurology, University of Eastern Finland and Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Jussi Paananen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Tiina Laiterä
- Institute of Clinical Medicine -Neurosurgery, University of Eastern Finland and Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland
| | - Maria Kojoukhova
- Institute of Clinical Medicine -Neurosurgery, University of Eastern Finland and Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland
| | - Anna Sutela
- Institute of Clinical Medicine - Pathology, University of Eastern Finland and Department of Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Ritva Vanninen
- Institute of Clinical Medicine - Pathology, University of Eastern Finland and Department of Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Marjo Laitinen
- Institute of Clinical Medicine -Neurology, University of Eastern Finland and Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Tuomas Rauramaa
- Institute of Clinical Medicine - Radiology, University of Eastern Finland and Department of Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Anne M Koivisto
- Institute of Clinical Medicine -Neurology, University of Eastern Finland and Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Anne M Remes
- Medical Research Center, Oulu University Hospital, Oulu, Finland and Research Unit of Clinical Neuroscience, Neurology, University of Oulu, Oulu, Finland
| | - Hilkka Soininen
- Institute of Clinical Medicine -Neurology, University of Eastern Finland and Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Mitja Kurki
- Institute of Clinical Medicine -Neurosurgery, University of Eastern Finland and Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland.,Analytical and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, USA; Stanley Center for Psychiatric Research, Broad Institute for Harvard and MIT, USA
| | - Annakaisa Haapasalo
- Institute of Clinical Medicine -Neurology, University of Eastern Finland and Department of Neurology, Kuopio University Hospital, Kuopio, Finland.,A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Juha E Jääskeläinen
- Institute of Clinical Medicine -Neurosurgery, University of Eastern Finland and Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Clinical Medicine -Neurology, University of Eastern Finland and Department of Neurology, Kuopio University Hospital, Kuopio, Finland.,Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Ville Leinonen
- Institute of Clinical Medicine -Neurosurgery, University of Eastern Finland and Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
57
|
Barrera J, Subramanian S, Chiba-Falek O. Probing the role of PPARγ in the regulation of late-onset Alzheimer's disease-associated genes. PLoS One 2018; 13:e0196943. [PMID: 29723294 PMCID: PMC5933777 DOI: 10.1371/journal.pone.0196943] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/23/2018] [Indexed: 12/21/2022] Open
Abstract
Peroxisome proliferator-activated receptor-γ (PPARγ), is a transcription factor that governs pathways, such as lipid metabolism and immune response, that have been implicated in the etiology of LOAD. Previously, we established HepG2-derived cell-lines with stable knockdown of PPARγ gene, and showed an increase in mRNA levels of genes mapped in the APOE linkage disequilibrium (LD) region on chromosome 19q13.32, with the greatest effect observed for APOE-mRNA. Here, we extended the analysis using our PPARγ knockdown model system and investigated the broader effect on expression changes of genes implicated in LOAD via genome wide association studies (GWAS). We applied the nCounter gene expression assay (NanoString) using a panel of twenty-four LOAD-associated genes inferred by proximity to the top significantly associated SNPs. Two independent PPARγ knockdown cell-lines showed changes in mRNA levels of a total of seven genes compared to a control HepG2 cell-line; six of which, ABCA7, APOE, CASS4, CELF1, PTK2B, and ZCWPW1, were upregulated and one, DSG2, was downregulated upon PPARγ knockdown. Our results propose that PPARγ may act as a master regulator of the transcription of several genes involved in LOAD pathogenesis. Our study provided the premise for further analyses including a larger set of genes positioned within a wider range of linkage disequilibrium (LD) regions tagged by all LOAD significantly associated SNPs.
Collapse
Affiliation(s)
- Julio Barrera
- Department of Neurology, Duke University Medical Center, Durham, North Carolina, United States of America
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Shobana Subramanian
- Department of Neurology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Ornit Chiba-Falek
- Department of Neurology, Duke University Medical Center, Durham, North Carolina, United States of America
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
58
|
Fernández MV, Budde J, Del-Aguila JL, Ibañez L, Deming Y, Harari O, Norton J, Morris JC, Goate AM, NIA-LOAD family study group, NCRAD, Cruchaga C. Evaluation of Gene-Based Family-Based Methods to Detect Novel Genes Associated With Familial Late Onset Alzheimer Disease. Front Neurosci 2018; 12:209. [PMID: 29670507 PMCID: PMC5893779 DOI: 10.3389/fnins.2018.00209] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 03/15/2018] [Indexed: 12/22/2022] Open
Abstract
Gene-based tests to study the combined effect of rare variants on a particular phenotype have been widely developed for case-control studies, but their evolution and adaptation for family-based studies, especially studies of complex incomplete families, has been slower. In this study, we have performed a practical examination of all the latest gene-based methods available for family-based study designs using both simulated and real datasets. We examined the performance of several collapsing, variance-component, and transmission disequilibrium tests across eight different software packages and 22 models utilizing a cohort of 285 families (N = 1,235) with late-onset Alzheimer disease (LOAD). After a thorough examination of each of these tests, we propose a methodological approach to identify, with high confidence, genes associated with the tested phenotype and we provide recommendations to select the best software and model for family-based gene-based analyses. Additionally, in our dataset, we identified PTK2B, a GWAS candidate gene for sporadic AD, along with six novel genes (CHRD, CLCN2, HDLBP, CPAMD8, NLRP9, and MAS1L) as candidate genes for familial LOAD.
Collapse
Affiliation(s)
- Maria V. Fernández
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - John Budde
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Jorge L. Del-Aguila
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Laura Ibañez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Yuetiva Deming
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Oscar Harari
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Joanne Norton
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - John C. Morris
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Alison M. Goate
- Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | | | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
59
|
林 芳, 刘 鑫, 谢 婧, 罗 静, 奉 夏, 侯 德. [Verification of a sporadic Alzheimer disease model in SORL1 gene knockout mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:289-295. [PMID: 29643034 PMCID: PMC6744166 DOI: 10.3969/j.issn.1673-4254.2018.03.08] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To compare the behavioral and pathological features of SORL1 gene knockout mice with those of normal mice and APP/PSE1 mice to verify the feasibility of using SORL1 knockout mice as a model of sporadic Alzheimer disease. METHODS SORL1 gene of fertilized mouse eggs were edited using Crispr/Case9 technique. SORL1-/- mice were screened and identified by detecting the DNA sequence, and Western blotting was used to detect the expression of SORL1. SORL1-/- mice, control mice and APP/PSE1 mice all underwent Morris water maze test to assess their learning and memory abilities with positioning navigation and space exploration experiments. The expression of APP and Aβ in the brain of the mice was detected using immunohistochemistry and Western blotting, respectively. RESULTS DNA sequencing showed CAAT deletion in SORL1 gene in two chromosomes of SORL1-/- mice, and the control mice had intact SORL1 gene without the deletion; Western blotting did not detect the expression of the SORL1 in the brain of SORL1-/- mice. Morris water maze test showed that in positioning navigation experiment, the average avoidance latency was similar between SORL1-/- mice and APP/PSE1 mice (P>0.05) but increased significantly in both mice as compared with the control group (P<0.05); similar results were obtained in the space exploration experiment. Immunohistochemistry and Western blotting revealed significantly increased APP and Aβ expression in the brain tissue of both SORL1-/- mice and APP/PSE1 mice compared with the control mice without significant differences between the two transgenic mice. CONCLUSION SORL1-/- mice exhibit similar behavioral and pathological changes with APP/PSE1 mice and can be used as a model of sporadic Alzheimer's disease.
Collapse
Affiliation(s)
- 芳波 林
- />中南大学湘雅三医院神经内科,湖南 长沙 410013Department of Neurology, Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - 鑫 刘
- />中南大学湘雅三医院神经内科,湖南 长沙 410013Department of Neurology, Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - 婧雯 谢
- />中南大学湘雅三医院神经内科,湖南 长沙 410013Department of Neurology, Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - 静 罗
- />中南大学湘雅三医院神经内科,湖南 长沙 410013Department of Neurology, Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - 夏露 奉
- />中南大学湘雅三医院神经内科,湖南 长沙 410013Department of Neurology, Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - 德仁 侯
- />中南大学湘雅三医院神经内科,湖南 长沙 410013Department of Neurology, Third Xiangya Hospital of Central South University, Changsha 410013, China
| |
Collapse
|
60
|
林 芳, 刘 鑫, 谢 婧, 罗 静, 奉 夏, 侯 德. [Verification of a sporadic Alzheimer disease model in SORL1 gene knockout mice]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:289-295. [PMID: 29643034 PMCID: PMC6744166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Indexed: 10/15/2023]
Abstract
OBJECTIVE To compare the behavioral and pathological features of SORL1 gene knockout mice with those of normal mice and APP/PSE1 mice to verify the feasibility of using SORL1 knockout mice as a model of sporadic Alzheimer disease. METHODS SORL1 gene of fertilized mouse eggs were edited using Crispr/Case9 technique. SORL1-/- mice were screened and identified by detecting the DNA sequence, and Western blotting was used to detect the expression of SORL1. SORL1-/- mice, control mice and APP/PSE1 mice all underwent Morris water maze test to assess their learning and memory abilities with positioning navigation and space exploration experiments. The expression of APP and Aβ in the brain of the mice was detected using immunohistochemistry and Western blotting, respectively. RESULTS DNA sequencing showed CAAT deletion in SORL1 gene in two chromosomes of SORL1-/- mice, and the control mice had intact SORL1 gene without the deletion; Western blotting did not detect the expression of the SORL1 in the brain of SORL1-/- mice. Morris water maze test showed that in positioning navigation experiment, the average avoidance latency was similar between SORL1-/- mice and APP/PSE1 mice (P>0.05) but increased significantly in both mice as compared with the control group (P<0.05); similar results were obtained in the space exploration experiment. Immunohistochemistry and Western blotting revealed significantly increased APP and Aβ expression in the brain tissue of both SORL1-/- mice and APP/PSE1 mice compared with the control mice without significant differences between the two transgenic mice. CONCLUSION SORL1-/- mice exhibit similar behavioral and pathological changes with APP/PSE1 mice and can be used as a model of sporadic Alzheimer's disease.
Collapse
Affiliation(s)
- 芳波 林
- />中南大学湘雅三医院神经内科,湖南 长沙 410013Department of Neurology, Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - 鑫 刘
- />中南大学湘雅三医院神经内科,湖南 长沙 410013Department of Neurology, Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - 婧雯 谢
- />中南大学湘雅三医院神经内科,湖南 长沙 410013Department of Neurology, Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - 静 罗
- />中南大学湘雅三医院神经内科,湖南 长沙 410013Department of Neurology, Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - 夏露 奉
- />中南大学湘雅三医院神经内科,湖南 长沙 410013Department of Neurology, Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - 德仁 侯
- />中南大学湘雅三医院神经内科,湖南 长沙 410013Department of Neurology, Third Xiangya Hospital of Central South University, Changsha 410013, China
| |
Collapse
|
61
|
Vardarajan BN, Barral S, Jaworski J, Beecham GW, Blue E, Tosto G, Reyes‐Dumeyer D, Medrano M, Lantigua R, Naj A, Thornton T, DeStefano A, Martin E, Wang L, Brown L, Bush W, van Duijn C, Goate A, Farrer L, Haines JL, Boerwinkle E, Schellenberg G, Wijsman E, Pericak‐Vance MA, Mayeux R, The Alzheimer's Disease Sequencing Project MosleyCantwellLauraChildressMicahChouYi‐FanCweibelRebeccaGangadharanPrabhakaranKuzmaAmandaLinHan‐JenMalamonJohnMlynarskiElisabethNajAdamQuLimingSchellenbergGerardValladaresOttoWangWeixinZhangNancyBelowBoerwinkleEricBresslerJanFornageMyriamJianXueqiuLiuXiaomingBisBlueElizabethBrownLisaDayTylerDorschnerMichaelNafikovNavasPatNguyenHiepPsatyBruceRiceKennethSaadMohamadSohiHarkiratThorntonTimothyTsuangDebbyWangBowenWijsmanEllenAppelbaumElizabethCruchagaCarlosKoboldtDaniel CWaligorskiJason, Wang LS. Whole genome sequencing of Caribbean Hispanic families with late-onset Alzheimer's disease. Ann Clin Transl Neurol 2018; 5:406-417. [PMID: 29688227 PMCID: PMC5899906 DOI: 10.1002/acn3.537] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 12/15/2017] [Accepted: 12/20/2017] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE To identify rare causal variants underlying known loci that segregate with late-onset Alzheimer's disease (LOAD) in multiplex families. METHODS We analyzed whole genome sequences (WGS) from 351 members of 67 Caribbean Hispanic (CH) families from Dominican Republic and New York multiply affected by LOAD. Members of 67 CH and additional 47 Caucasian families underwent WGS as a part of the Alzheimer's Disease Sequencing Project (ADSP). All members of 67 CH families, an additional 48 CH families and an independent CH case-control cohort were subsequently genotyped for validation. Patients met criteria for LOAD, and controls were determined to be dementia free. We investigated rare variants segregating within families and gene-based associations with disease within LOAD GWAS loci. RESULTS A variant in AKAP9, p.R434W, segregated significantly with LOAD in two large families (OR = 5.77, 95% CI: 1.07-30.9, P = 0.041). In addition, missense mutations in MYRF and ASRGL1 under previously reported linkage peaks at 7q14.3 and 11q12.3 segregated completely in one family and in follow-up genotyping both were nominally significant (P < 0.05). We also identified rare variants in a number of genes associated with LOAD in prior genome wide association studies, including CR1 (P = 0.049), BIN1 (P = 0.0098) and SLC24A4 (P = 0.040). CONCLUSIONS AND RELEVANCE Rare variants in multiple genes influence the risk of LOAD disease in multiplex families. These results suggest that rare variants may underlie loci identified in genome wide association studies.
Collapse
Affiliation(s)
- Badri N. Vardarajan
- The Taub Institute for Research on Alzheimer's Disease and the Aging BrainNew York,The Gertrude H. Sergievsky CenterColumbia UniversityThe New York Presbyterian HospitalNew YorkNew York,Department of Systems BiologyColumbia UniversityThe New York Presbyterian HospitalNew YorkNew York
| | - Sandra Barral
- The Taub Institute for Research on Alzheimer's Disease and the Aging BrainNew York,The Gertrude H. Sergievsky CenterColumbia UniversityThe New York Presbyterian HospitalNew YorkNew York,The Department of NeurologyColumbia UniversityThe New York Presbyterian HospitalNew YorkNew York
| | - James Jaworski
- Dr. John T. Macdonald Foundation Department of Human GeneticsThe John P. Hussman Institute for Human GenomicsMiamiFlorida
| | - Gary W. Beecham
- Dr. John T. Macdonald Foundation Department of Human GeneticsThe John P. Hussman Institute for Human GenomicsMiamiFlorida
| | - Elizabeth Blue
- Division of Medical GeneticsDepartment of MedicineUniversity of WashingtonSeattleWashington
| | - Giuseppe Tosto
- The Taub Institute for Research on Alzheimer's Disease and the Aging BrainNew York,The Gertrude H. Sergievsky CenterColumbia UniversityThe New York Presbyterian HospitalNew YorkNew York
| | - Dolly Reyes‐Dumeyer
- The Taub Institute for Research on Alzheimer's Disease and the Aging BrainNew York,The Gertrude H. Sergievsky CenterColumbia UniversityThe New York Presbyterian HospitalNew YorkNew York
| | - Martin Medrano
- School of MedicineMother and Teacher Pontifical Catholic UniversitySantiagoDominican Republic
| | - Rafael Lantigua
- The Taub Institute for Research on Alzheimer's Disease and the Aging BrainNew York,Department of MedicineColumbia UniversityThe New York Presbyterian HospitalNew YorkNew York
| | - Adam Naj
- School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Timothy Thornton
- Department of BiostatisticsUniversity of WashingtonSeattleWashington
| | | | - Eden Martin
- Dr. John T. Macdonald Foundation Department of Human GeneticsThe John P. Hussman Institute for Human GenomicsMiamiFlorida
| | - Li‐San Wang
- School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Lisa Brown
- Department of BiostatisticsUniversity of WashingtonSeattleWashington
| | - William Bush
- Department of Biostatistics and EpidemiologyCase Western Reserve UniversityClevelandOhio
| | | | | | | | - Jonathan L. Haines
- Department of Biostatistics and EpidemiologyCase Western Reserve UniversityClevelandOhio
| | | | | | - Ellen Wijsman
- Division of Medical GeneticsDepartment of MedicineUniversity of WashingtonSeattleWashington,Department of BiostatisticsUniversity of WashingtonSeattleWashington
| | - Margaret A. Pericak‐Vance
- Dr. John T. Macdonald Foundation Department of Human GeneticsThe John P. Hussman Institute for Human GenomicsMiamiFlorida
| | - Richard Mayeux
- The Taub Institute for Research on Alzheimer's Disease and the Aging BrainNew York,The Gertrude H. Sergievsky CenterColumbia UniversityThe New York Presbyterian HospitalNew YorkNew York,Department of Systems BiologyColumbia UniversityThe New York Presbyterian HospitalNew YorkNew York,Department of PsychiatryColumbia UniversityThe New York Presbyterian HospitalNew YorkNew York,The Department of EpidemiologySchool of Public HealthColumbia UniversityNew YorkNew York
| | | | - Li-San Wang
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain New York.,The Gertrude H. Sergievsky Center Columbia University The New York Presbyterian Hospital New York New York.,Department of Systems Biology Columbia University The New York Presbyterian Hospital New York New York.,The Department of Neurology Columbia University The New York Presbyterian Hospital New York New York.,Dr. John T. Macdonald Foundation Department of Human Genetics The John P. Hussman Institute for Human Genomics Miami Florida.,Division of Medical Genetics Department of Medicine University of Washington Seattle Washington.,School of Medicine Mother and Teacher Pontifical Catholic University Santiago Dominican Republic.,Department of Medicine Columbia University The New York Presbyterian Hospital New York New York.,School of Medicine University of Pennsylvania Philadelphia Pennsylvania.,Department of Biostatistics University of Washington Seattle Washington.,Boston University School of Medicine Boston Massachusetts.,Department of Biostatistics and Epidemiology Case Western Reserve University Cleveland Ohio.,Erasmus University Medical Center Rotterdam Netherlands.,Mount Sinai School of Medicine New York New York.,University of Texas Houston Texas.,Department of Psychiatry Columbia University The New York Presbyterian Hospital New York New York.,The Department of Epidemiology School of Public Health Columbia University New York New York
| |
Collapse
|
62
|
Sao T, Yoshino Y, Yamazaki K, Ozaki Y, Mori Y, Ochi S, Yoshida T, Mori T, Iga JI, Ueno SI. MEF2C mRNA expression and cognitive function in Japanese patients with Alzheimer's disease. Psychiatry Clin Neurosci 2018; 72:160-167. [PMID: 29112298 DOI: 10.1111/pcn.12618] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/12/2017] [Accepted: 10/30/2017] [Indexed: 12/15/2022]
Abstract
AIM Despite continuing research into Alzheimer's disease (AD), its pathological mechanisms and modulating factors remain unknown. Several genes influence AD pathogenesis by affecting inflammatory pathways. Myocyte-enhancer factor 2C (MEF2C) is one such candidate gene for AD. METHODS We examined MEF2C mRNA expression levels and methylation rates of CpG on its promoter region in peripheral leukocytes from Japanese AD patients compared with age- and sex-matched control subjects. RESULTS In peripheral leukocytes, MEF2C mRNA expression levels in AD subjects were significantly lower than those in control subjects (0.86 ± 0.25 vs 0.99 ± 0.27, respectively, P = 0.007) and were correlated with the Alzheimer's Disease Assessment Scale (r = -0.345, P = 0.049) and the Mini Mental State Examination (r = 0.324, P = 0.02). No significant differences were found in methylation rates between AD and control subjects. CONCLUSION MEF2C mRNA expression in leukocytes may be a biological marker for cognitive decline in AD.
Collapse
Affiliation(s)
- Tomoko Sao
- Department of Neuropsychiatry, Molecules, and Function, Ehime University Graduate School of Medicine, Toon, Japan
| | - Yuta Yoshino
- Department of Neuropsychiatry, Molecules, and Function, Ehime University Graduate School of Medicine, Toon, Japan
| | - Kiyohiro Yamazaki
- Department of Neuropsychiatry, Molecules, and Function, Ehime University Graduate School of Medicine, Toon, Japan
| | - Yuki Ozaki
- Department of Neuropsychiatry, Molecules, and Function, Ehime University Graduate School of Medicine, Toon, Japan
| | - Yoko Mori
- Department of Neuropsychiatry, Molecules, and Function, Ehime University Graduate School of Medicine, Toon, Japan
| | - Shinichiro Ochi
- Department of Neuropsychiatry, Molecules, and Function, Ehime University Graduate School of Medicine, Toon, Japan
| | - Taku Yoshida
- Department of Neuropsychiatry, Molecules, and Function, Ehime University Graduate School of Medicine, Toon, Japan
| | - Takaaki Mori
- Department of Neuropsychiatry, Molecules, and Function, Ehime University Graduate School of Medicine, Toon, Japan
| | - Jun-Ichi Iga
- Department of Neuropsychiatry, Molecules, and Function, Ehime University Graduate School of Medicine, Toon, Japan
| | - Shu-Ichi Ueno
- Department of Neuropsychiatry, Molecules, and Function, Ehime University Graduate School of Medicine, Toon, Japan
| |
Collapse
|
63
|
Young JE, Fong LK, Frankowski H, Petsko GA, Small SA, Goldstein LSB. Stabilizing the Retromer Complex in a Human Stem Cell Model of Alzheimer's Disease Reduces TAU Phosphorylation Independently of Amyloid Precursor Protein. Stem Cell Reports 2018; 10:1046-1058. [PMID: 29503090 PMCID: PMC5919412 DOI: 10.1016/j.stemcr.2018.01.031] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 01/25/2018] [Accepted: 01/26/2018] [Indexed: 12/22/2022] Open
Abstract
Developing effective therapeutics for complex diseases such as late-onset, sporadic Alzheimer’s disease (SAD) is difficult due to genetic and environmental heterogeneity in the human population and the limitations of existing animal models. Here, we used hiPSC-derived neurons to test a compound that stabilizes the retromer, a highly conserved multiprotein assembly that plays a pivotal role in trafficking molecules through the endosomal network. Using this human-specific system, we have confirmed previous data generated in murine models and show that retromer stabilization has a potentially beneficial effect on amyloid beta generation from human stem cell-derived neurons. We further demonstrate that manipulation of retromer complex levels within neurons affects pathogenic TAU phosphorylation in an amyloid-independent manner. Taken together, our work demonstrates that retromer stabilization is a promising candidate for therapeutic development in AD and highlights the advantages of testing novel compounds in a human-specific, neuronal system. A retromer stabilizing molecule reduces Aβ and phospho-TAU levels in human neurons The molecule reduces Aβ and pTau in both SAD and FAD cell lines Retromer stabilization reduces tau phosphorylation in an APP-independent manner Confirms studies in mice and highlights hiPSCs as a preclinical model
Collapse
Affiliation(s)
- Jessica E Young
- Department of Pathology, University of Washington, Seattle, WA 98109, USA; Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA.
| | - Lauren K Fong
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA
| | - Harald Frankowski
- Department of Pathology, University of Washington, Seattle, WA 98109, USA; Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Gregory A Petsko
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medical College, New York, NY 10021, USA
| | - Scott A Small
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Lawrence S B Goldstein
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, CA 92093, USA.
| |
Collapse
|
64
|
INPP5D rs35349669 polymorphism with late-onset Alzheimer's disease: A replication study and meta-analysis. Oncotarget 2018; 7:69225-69230. [PMID: 27750211 PMCID: PMC5342472 DOI: 10.18632/oncotarget.12648] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 10/02/2016] [Indexed: 11/25/2022] Open
Abstract
Inositol polyphosphate-5-phosphatase (INPP5D) was reported to be associated with Alzheimer's disease (AD) through modulating the inflammatory process and immune response. A recent genome-wide association study discovered a new locus single nucleotide polymorphism (SNP, rs35349669) of INPP5D which was significantly associated with susceptibility to late-onset Alzheimer's disease (LOAD) in Caucasians. In this study, we investigated the relations between the INPP5D polymorphism rs35349669 and LOAD in Han Chinese population comprising 984 LOAD cases and 1352 healthy controls being matched for age and gender. Our results showed no obvious differences in the genotypic or allelic distributions of rs35349669 polymorphism between LOAD cases and healthy controls (genotype: p = 0.167; allele: p = 0.094). Additionally, when these data were stratified by APOEε4 status, there are still no evident differences in the genotypic or allelic distributions in APOEε4 carriers (p > 0.05). Furthermore, meta-analysis of 81964 individuals confirmed that rs35349669 was significantly associated with the risk for LOAD (OR=1.08, 95%CI=1.06-1.11), but the results remained negative in Chinese subgroup (OR=0.77, 95%CI=0.53-1.13). Overall, the current evidence did not indicate that INPP5D rs35349669 polymorphism play a role in the genetic predisposition to LOAD in Chinese population.
Collapse
|
65
|
Chung J, Zhang X, Allen M, Wang X, Ma Y, Beecham G, Montine TJ, Younkin SG, Dickson DW, Golde TE, Price ND, Ertekin-Taner N, Lunetta KL, Mez J, Alzheimer’s Disease Genetics Consortium, Mayeux R, Haines JL, Pericak-Vance MA, Schellenberg G, Jun GR, Farrer LA. Genome-wide pleiotropy analysis of neuropathological traits related to Alzheimer's disease. Alzheimers Res Ther 2018; 10:22. [PMID: 29458411 PMCID: PMC5819208 DOI: 10.1186/s13195-018-0349-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 01/22/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Simultaneous consideration of two neuropathological traits related to Alzheimer's disease (AD) has not been attempted in a genome-wide association study. METHODS We conducted genome-wide pleiotropy analyses using association summary statistics from the Beecham et al. study (PLoS Genet 10:e1004606, 2014) for AD-related neuropathological traits, including neuritic plaque (NP), neurofibrillary tangle (NFT), and cerebral amyloid angiopathy (CAA). Significant findings were further examined by expression quantitative trait locus and differentially expressed gene analyses in AD vs. control brains using gene expression data. RESULTS Genome-wide significant pleiotropic associations were observed for the joint model of NP and NFT (NP + NFT) with the single-nucleotide polymorphism (SNP) rs34487851 upstream of C2orf40 (alias ECRG4, P = 2.4 × 10-8) and for the joint model of NFT and CAA (NFT + CAA) with the HDAC9 SNP rs79524815 (P = 1.1 × 10-8). Gene-based testing revealed study-wide significant associations (P ≤ 2.0 × 10-6) for the NFT + CAA outcome with adjacent genes TRAPPC12, TRAPPC12-AS1, and ADI1. Risk alleles of proxy SNPs for rs79524815 were associated with significantly lower expression of HDAC9 in the brain (P = 3.0 × 10-3), and HDAC9 was significantly downregulated in subjects with AD compared with control subjects in the prefrontal (P = 7.9 × 10-3) and visual (P = 5.6 × 10-4) cortices. CONCLUSIONS Our findings suggest that pleiotropy analysis is a useful approach to identifying novel genetic associations with complex diseases and their endophenotypes. Functional studies are needed to determine whether ECRG4 or HDAC9 is plausible as a therapeutic target.
Collapse
Affiliation(s)
- Jaeyoon Chung
- Bioinformatics Graduate Program, Boston University, Boston, MA USA
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA USA
| | - Xiaoling Zhang
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA USA
| | - Mariet Allen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL USA
| | - Xue Wang
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL USA
| | - Yiyi Ma
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA USA
| | - Gary Beecham
- Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL USA
| | | | | | | | - Todd E. Golde
- Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL USA
| | - Nathan D. Price
- Institute for Systems Biology, University of Washington, Seattle, WA USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL USA
| | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Jesse Mez
- Department of Neurology, Boston University School of Medicine, Boston, MA USA
| | - Alzheimer’s Disease Genetics Consortium
- Bioinformatics Graduate Program, Boston University, Boston, MA USA
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA USA
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL USA
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL USA
- Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL USA
- Department of Pathology, University of Washington, Seattle, WA USA
- Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL USA
- Institute for Systems Biology, University of Washington, Seattle, WA USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
- Department of Neurology, Boston University School of Medicine, Boston, MA USA
- Department of Neurology and Sergievsky Center, Columbia University, New York, NY USA
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA USA
- Neurogenetics and Integrated Genomics, Andover Innovative Medicines Institute, Eisai Inc., Andover, MA USA
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA USA
| | - Richard Mayeux
- Department of Neurology and Sergievsky Center, Columbia University, New York, NY USA
| | - Jonathan L. Haines
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH USA
| | | | - Gerard Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Gyungah R. Jun
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA USA
- Neurogenetics and Integrated Genomics, Andover Innovative Medicines Institute, Eisai Inc., Andover, MA USA
| | - Lindsay A. Farrer
- Bioinformatics Graduate Program, Boston University, Boston, MA USA
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, MA USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
- Department of Neurology, Boston University School of Medicine, Boston, MA USA
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA USA
| |
Collapse
|
66
|
Claes C, Van den Daele J, Verfaillie CM. Generating tissue-resident macrophages from pluripotent stem cells: Lessons learned from microglia. Cell Immunol 2018; 330:60-67. [PMID: 29433896 DOI: 10.1016/j.cellimm.2018.01.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/31/2018] [Accepted: 01/31/2018] [Indexed: 02/07/2023]
Abstract
Over the past decades, the importance of the immune system in a broad scope of pathologies, has drawn attention towards tissue-resident macrophages, such as microglia in the brain. To enable the study of for instance microglia, it is crucial to recreate in vitro (and in vivo) assays. However, very fast loss of tissue-specific features of primary tissue resident macrophages, including microglia, upon in vitro culture has complicated such studies. Moreover, limited knowledge of macrophage developmental pathways and the role of local 'niche factors', has hampered the generation of tissue-resident macrophages from pluripotent stem cells (PSC). Recent data on the ontogeny of tissue-resident macrophages, combined with bulk and single cell RNAseq studies have identified the distinct origins and gene profile of microglia compared to other myeloid cells. As a result, over the past years, protocols have been published to create hPSC-derived microglia-'like' cells, as these cells are considered potential new therapeutic targets for therapies to treat neurodegenerative diseases. In this review we will provide an overview of different approaches taken to generate human microglia in vitro, taking into account their origin, and resemblance to their in vivo counterpart. Finally, we will discuss cell-extrinsic (culture conditions) and intrinsic factors (transcriptional machinery and epigenetics) that we believe can improve future differentiation protocols of tissue-resident macrophages from stem cells.
Collapse
Affiliation(s)
- Christel Claes
- Department of Development and Regeneration, Stem Cell and Developmental Biology, Stem Cell Institute Leuven, KU Leuven, Leuven, Belgium
| | - Johanna Van den Daele
- Department of Development and Regeneration, Stem Cell and Developmental Biology, Stem Cell Institute Leuven, KU Leuven, Leuven, Belgium
| | - Catherine M Verfaillie
- Department of Development and Regeneration, Stem Cell and Developmental Biology, Stem Cell Institute Leuven, KU Leuven, Leuven, Belgium.
| |
Collapse
|
67
|
Taipa R, Sousa AL, Melo Pires M, Sousa N. Does the Interplay Between Aging and Neuroinflammation Modulate Alzheimer's Disease Clinical Phenotypes? A Clinico-Pathological Perspective. J Alzheimers Dis 2018; 53:403-17. [PMID: 27176075 DOI: 10.3233/jad-160121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder and is the most common cause of dementia worldwide. Cumulative data suggests that neuroinflammation plays a prominent and early role in AD, and there is compelling data from different research groups of age-associated dysregulation of the neuroimmune system. From the clinical point of view, despite clinical resemblance and neuropathological findings, there are important differences between the group of patients with sporadic early-onset (<65 years old) and late-onset AD (>65 years old). Thus, it seems important to understand the age-dependent relationship between neuroinflammation and the underlying biology of AD in order to identify potential explanations for clinical heterogeneity, interpret biomarkers, and promote the best treatment to different clinical AD phenotypes. The study of the delicate balance between pro-inflammatory or anti-inflammatory sides of immune players in the different ages of onset of AD would be important to understand treatment efficacy in clinical trials and eventually, not only direct treatment to early disease stages, but also the possibility of establishing different treatment approaches depending on the age of the patient. In this review, we would like to summarize what is currently known about the interplay between "normal" age associated inflammatory changes and AD pathological mechanisms, and also the potential differences between early-onset and late-onset AD taking into account the age-related neuroimmune background at disease onset.
Collapse
Affiliation(s)
- Ricardo Taipa
- Neuropathology Unit, Department of Neuroscience, Hospital Santo António - Centro Hospitalar do Porto, Porto, Portugal.,Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - Ana Luísa Sousa
- Department of Neurology, Hospital Santo António - Centro Hospitalar do Porto, Porto, Portugal
| | - Manuel Melo Pires
- Neuropathology Unit, Department of Neuroscience, Hospital Santo António - Centro Hospitalar do Porto, Porto, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| |
Collapse
|
68
|
ZCWPW1 is associated with late-onset Alzheimer's disease in Han Chinese: a replication study and meta-analyses. Oncotarget 2018; 7:20305-11. [PMID: 26958812 PMCID: PMC4991456 DOI: 10.18632/oncotarget.7945] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 02/09/2016] [Indexed: 01/22/2023] Open
Abstract
Recently, a large genome-wide association study (GWAS) has identified a novel variant (rs1476679) within ZCWPW1 showing strong association with late-onset Alzheimer's disease (LOAD) in Caucasian. However, the effect of rs1476679 on other populations remains unclear. In order to explore whether rs1476679 is also associated with the LOAD risk in other ethnic groups, we recruited 2350 unrelated Northern Han Chinese subjects, which include 992 LOAD patients and 1358 healthy controls. Analysis of data from these subjects suggests that the rs1476679 polymorphism is significantly associated with the LOAD (genotype P = 0.017, allele P = 0.044). The logistic regression reveals the C allele at rs1476679 is a protective factor for LOAD in the dominant model (OR = 0.779, 95%CI = 0.659–0.921, Pc = 0.009) adjusting for gender, age and APOE ε4 status. Furthermore, rs1476679 can decrease the AD risk (Dominant: OR = 0.733, 95%CI = 0.607–0.884, Pc = 0.006; Additive: OR = 0.820, 95%CI = 0.708–0.950, Pc = 0.048) in APOE ε4 non-carriers after stratification. Furthermore, meta-analysis of 82525 individuals confirmed that rs1476679 within ZCWPW1 decreased the risk of LOAD (OR = 0.91, 95%CI = 0.89–0.94). To summarize, the rs1476679 polymorphism in ZCWPW1 is associated with LOAD in Northern Han Chinese population.
Collapse
|
69
|
Wood PL, Cebak JE, Woltjer RL. Diacylglycerols as biomarkers of sustained immune activation in Proteinopathies associated with dementia. Clin Chim Acta 2018; 476:107-110. [DOI: 10.1016/j.cca.2017.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/10/2017] [Accepted: 11/12/2017] [Indexed: 12/12/2022]
|
70
|
Ramanan VK, Przybelski SA, Graff-Radford J, Castillo AM, Lowe VJ, Mielke MM, Roberts RO, Reid RI, Knopman DS, Jack CR, Petersen RC, Vemuri P. Statins and Brain Health: Alzheimer's Disease and Cerebrovascular Disease Biomarkers in Older Adults. J Alzheimers Dis 2018; 65:1345-1352. [PMID: 30149450 PMCID: PMC6260813 DOI: 10.3233/jad-180446] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Statins have been proposed to reduce the risk of Alzheimer's disease (AD). OBJECTIVE Assess whether long-term statin use was associated with neuroimaging biomarkers of aging and dementia. METHODS Methods: We analyzed neuroimaging biomarkers in 1,160 individuals aged 65+ from the Mayo Clinic Study of Aging, a population-based prospective longitudinal study of cognitive aging. RESULTS Statin-treated (5+ years of therapy) individuals had greater burden of mid-and late-life cardiovascular disease (p < 0.001) than statin-untreated (≤3 months) individuals. Lower fractional anisotropy in the genu of the corpus callosum, an early marker of cerebrovascular disease, was associated with long-term statin exposure (p < 0.035). No significant associations were identified between long-term statin exposure and cerebral amyloid or tau burden, AD pattern neurodegeneration, or white matter hyperintensity burden. CONCLUSIONS Long-term statin therapy was not associated with differences in AD biomarkers. Individuals with long-term statin exposure had worse white matter integrity in the genu of the corpus callosum, consistent with the coexistence of higher cerebrovascular risk factor burden in this group.
Collapse
Affiliation(s)
- Vijay K Ramanan
- Department of Neurology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Scott A. Przybelski
- Department of Health Sciences Research, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | | | - Anna M. Castillo
- Department of Health Sciences Research, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Michelle M. Mielke
- Department of Neurology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
- Department of Health Sciences Research, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Rosebud O. Roberts
- Department of Neurology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
- Department of Health Sciences Research, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Robert I. Reid
- Department of Information Technology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - David S. Knopman
- Department of Neurology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Clifford R. Jack
- Department of Radiology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Ronald C. Petersen
- Department of Neurology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
- Department of Health Sciences Research, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| |
Collapse
|
71
|
Liu SL, Wang XC, Tan MS, Wang HF, Zhang W, Wang ZX, Yu JT, Tan L. NME8 rs2718058 polymorphism with Alzheimer's disease risk: a replication and meta-analysis. Oncotarget 2017; 7:36014-36020. [PMID: 27144521 PMCID: PMC5094979 DOI: 10.18632/oncotarget.9086] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/11/2016] [Indexed: 01/28/2023] Open
Abstract
Recently, a large meta-analysis of five genome wide association studies (GWAS) has identified that a novel single nucleotide polymorphism (SNP) rs2718058, adjacent to gene NME8 on chromosome 7p14.1, was associated with late-onset Alzheimer's disease (LOAD) in Caucasians. However, the effect of rs2718058 on other populations remains unclear. In order to explore the relationship between rs2718058 and LOAD risk in a North Han Chinese population, we recruited 984 LOAD cases and 1354 healthy controls that matched for sex and age in this study. The results showed no significant differences in the genotypic or allelic distributions of rs2718058 polymorphism between LOAD cases and healthy controls, even though after stratification for APOE ε4 status and statistical adjustment for age, gender and APOE ε4 status (p > 0.05). However, a meta-analysis conducted in a sample of 82513 individuals confirmed a significant association between SNP rs2718058 and LOAD risk (OR = 1.08, 95%CI = 1.05-1.11) in the whole population. But there was still no positive results in Chinese subgroup (OR = 1.05, 95%CI = 0.93-1.17). In conclusion, the rs2718058 near gene NME8 on chromosome 7p14.1 might not play a major role in the genetic predisposition to LOAD in the North Han Chinese.
Collapse
Affiliation(s)
- Shu-Lei Liu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, PR China
| | - Xue-Chun Wang
- Department of Radiology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, PR China
| | - Meng-Shan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, PR China
| | - Hui-Fu Wang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, PR China
| | - Wei Zhang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, PR China
| | - Zi-Xuan Wang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, PR China
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, PR China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, PR China
| |
Collapse
|
72
|
Bhatia S, Fu Y, Hsiao JHT, Halliday GM, Kim WS. Deletion of Alzheimer's Disease Risk Gene ABCA7 Alters White Adipose Tissue Development and Leptin Levels. J Alzheimers Dis Rep 2017; 1:237-247. [PMID: 30480241 PMCID: PMC6159609 DOI: 10.3233/adr-170029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
ATP-binding cassette A7 (ABCA7) is a genetic risk factor for late-onset Alzheimer’s disease (AD). It belongs to a group of transporter genes that specializes in regulating lipid transport in the periphery as well as in the brain. ABCA7 has been implicated in a number of roles relating to AD pathology, including phagocytic clearance of amyloid-β peptides. We have discovered that deletion of ABCA7 in mouse causes a dramatic reduction in white adipose tissue (WAT) in female mice. WAT is important in AD context because it is the primary producer of leptin, which is a hormone that is known to modulate AD neuropathology. WAT in male Abca7–/– mice was not altered. The pathological link between ABCA7 and WAT that impacts on AD is unknown. Our transcription analysis revealed that lipin-1 expression was significantly upregulated in female Abca7–/– mice, indicating that ABCA7 affects WAT development. The circulating leptin level was significantly reduced in female Abca7–/– mice without any change in WAT leptin mRNA or protein expression, indicating that ABCA7 does not affect leptin production, but alters the circulating leptin level indirectly by affecting WAT development. Insulin is a key hormone that regulates WAT development, i.e., adipogenesis, and it was significantly reduced in female Abca7–/– mice. These data when put together suggest that ABCA7 plays a role in regulating WAT development and consequently circulating leptin levels, which are known to modulate AD neuropathology.
Collapse
Affiliation(s)
- Surabhi Bhatia
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - YuHong Fu
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Jen-Hsiang T Hsiao
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia.,School of Medical Sciences, University of New South Wales and Neuroscience Research Australia, Randwick, NSW, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia.,School of Medical Sciences, University of New South Wales and Neuroscience Research Australia, Randwick, NSW, Australia
| | - Woojin Scott Kim
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia.,School of Medical Sciences, University of New South Wales and Neuroscience Research Australia, Randwick, NSW, Australia
| |
Collapse
|
73
|
Wright K, Bihaqi SW, Lahouel A, Masoud A, Mushtaq F, Leso A, Eid A, Zawia NH. Importance of tau in cognitive decline as revealed by developmental exposure to lead. Toxicol Lett 2017; 284:63-69. [PMID: 29203278 DOI: 10.1016/j.toxlet.2017.11.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 12/21/2022]
Abstract
Previous reports by us have determined that developmental exposure to the heavy metal lead (Pb) resulted in cognitive impairment in aging wildtype mice, and a latent induction in biomarkers associated with both the tau and amyloid pathways. However, the relationship between these two pathways and their correlation to cognitive performance needs to be scrutinized. Here, we investigated the impact of developmental Pb (0.2%) exposure on the amyloid and tau pathways in a transgenic mouse model lacking the tau gene. Cognitive function, and levels of intermediates in the amyloid and tau pathways following postnatal Pb exposure were assessed on young adult and mature transgenic mice. No significant difference in behavioral performance, amyloid precursor protein (APP), or amyloid beta (Aβ) levels was observed in transgenic mice exposed to Pb. Regulators of the tau pathway were impacted by the absence of tau, but no additional change was imparted by Pb exposure. These results revealed that developmental Pb exposure does not cause cognitive decline or change the expression of the amyloid pathway in the absence of tau. The essentiality of tau to mediate cognitive decline by environmental perturbations needs further investigation.
Collapse
Affiliation(s)
- K Wright
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston Rhode Island, 02881, USA
| | - S W Bihaqi
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston Rhode Island, 02881, USA
| | - A Lahouel
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston Rhode Island, 02881, USA
| | - A Masoud
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston Rhode Island, 02881, USA; Biochemical Technology Program, Faculty of Applied Science, Thamar University, Yemen
| | - F Mushtaq
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston Rhode Island, 02881, USA
| | - A Leso
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston Rhode Island, 02881, USA
| | - A Eid
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston Rhode Island, 02881, USA; Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston Rhode Island, 02881, USA
| | - N H Zawia
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston Rhode Island, 02881, USA; Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston Rhode Island, 02881, USA; Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston Rhode Island, 02881, USA.
| |
Collapse
|
74
|
Lerner R, Post JM, Ellis SR, Vos DRN, Heeren RMA, Lutz B, Bindila L. Simultaneous lipidomic and transcriptomic profiling in mouse brain punches of acute epileptic seizure model compared to controls. J Lipid Res 2017; 59:283-297. [PMID: 29208697 DOI: 10.1194/jlr.m080093] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/28/2017] [Indexed: 01/07/2023] Open
Abstract
In this study, we report the development of a dual extraction protocol for RNA and lipids, including phospholipids, endocannabinoids, and arachidonic acid, at high spatial resolution, e.g., brain punches obtained from whole frozen brains corresponding to four brain subregions: dorsal hippocampus, ventral hippocampus, basolateral amygdala, and hypothalamus. This extraction method combined with LC/multiple reaction monitoring for lipid quantifi-cation and quantitative PCR for RNA investigation allows lipidomic and transcriptomic profiling from submilligram amounts of tissue, thus benefiting the time and animal costs for analysis and the data reliability due to prevention of biological variability between animal batches and/or tissue heterogeneity, as compared with profiling in distinct animal batches. Moreover, the method allows a higher extraction efficiency and integrity preservation for RNA, while allowing concurrently quantitative analysis of low and high abundant lipids. The method was applied for brain punches obtained 1 h after kainic acid-induced epileptic seizures in mice (n = 10) compared with controls (n = 10), and enabled the provision of valuable new insights into the subregional lipid and RNA changes with epilepsy, highlighting its potential as a new viable tool in quantitative neurobiology.
Collapse
Affiliation(s)
- Raissa Lerner
- University Medical Center of Johannes Gutenberg University Mainz, Institute of Physiological Chemistry, 55128 Mainz, Germany; and
| | - Julia M Post
- University Medical Center of Johannes Gutenberg University Mainz, Institute of Physiological Chemistry, 55128 Mainz, Germany; and
| | - Shane R Ellis
- Maastricht MultiModal Molecular Imaging Institute (M4I), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - D R Naomi Vos
- Maastricht MultiModal Molecular Imaging Institute (M4I), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ron M A Heeren
- Maastricht MultiModal Molecular Imaging Institute (M4I), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Beat Lutz
- University Medical Center of Johannes Gutenberg University Mainz, Institute of Physiological Chemistry, 55128 Mainz, Germany; and
| | - Laura Bindila
- University Medical Center of Johannes Gutenberg University Mainz, Institute of Physiological Chemistry, 55128 Mainz, Germany; and
| |
Collapse
|
75
|
Deczkowska A, Matcovitch-Natan O, Tsitsou-Kampeli A, Ben-Hamo S, Dvir-Szternfeld R, Spinrad A, Singer O, David E, Winter DR, Smith LK, Kertser A, Baruch K, Rosenzweig N, Terem A, Prinz M, Villeda S, Citri A, Amit I, Schwartz M. Mef2C restrains microglial inflammatory response and is lost in brain ageing in an IFN-I-dependent manner. Nat Commun 2017; 8:717. [PMID: 28959042 PMCID: PMC5620041 DOI: 10.1038/s41467-017-00769-0] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 07/23/2017] [Indexed: 11/08/2022] Open
Abstract
During ageing, microglia acquire a phenotype that may negatively affect brain function. Here we show that ageing microglial phenotype is largely imposed by interferon type I (IFN-I) chronically present in aged brain milieu. Overexpression of IFN-β in the CNS of adult wild-type mice, but not of mice lacking IFN-I receptor on their microglia, induces an ageing-like transcriptional microglial signature, and impairs cognitive performance. Furthermore, we demonstrate that age-related IFN-I milieu downregulates microglial myocyte-specific enhancer factor 2C (Mef2C). Immune challenge in mice lacking Mef2C in microglia results in an exaggerated microglial response and has an adverse effect on mice behaviour. Overall, our data indicate that the chronic presence of IFN-I in the brain microenvironment, which negatively affects cognitive function, is mediated via modulation of microglial activity. These findings may shed new light on other neurological conditions characterized by elevated IFN-I signalling in the brain.Microglia cells in the brain regulate immune responses, but in ageing can negatively affect brain function. Here the authors show that the chronic presence of type I interferon in aged mouse brain impedes cognitive ability by altering microglia transcriptome and limiting Mef2C, a microglia 'off' signal.
Collapse
Affiliation(s)
| | - Orit Matcovitch-Natan
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | | | - Sefi Ben-Hamo
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Raz Dvir-Szternfeld
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Amit Spinrad
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Oded Singer
- Faculty of Biochemistry, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Deborah R Winter
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Lucas K Smith
- Department of Anatomy, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Alexander Kertser
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Kuti Baruch
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Neta Rosenzweig
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Anna Terem
- Department of Biological Chemistry, Institute of Life Sciences, Faculty of Natural Sciences, The Hebrew University, Jerusalem, 91904, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem, 91904, Israel
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, 79104, Germany
| | - Saul Villeda
- Department of Anatomy, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ami Citri
- Department of Biological Chemistry, Institute of Life Sciences, Faculty of Natural Sciences, The Hebrew University, Jerusalem, 91904, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University, Jerusalem, 91904, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| | - Michal Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| |
Collapse
|
76
|
Sharma N, Khurana N, Muthuraman A. Lower vertebrate and invertebrate models of Alzheimer's disease - A review. Eur J Pharmacol 2017; 815:312-323. [PMID: 28943103 DOI: 10.1016/j.ejphar.2017.09.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 08/20/2017] [Accepted: 09/13/2017] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease is a common neurodegenerative disorder which is characterized by the presence of beta- amyloid protein and neurofibrillary tangles (NFTs) in the brain. Till now, various higher vertebrate models have been in use to study the pathophysiology of this disease. But, these models possess some limitations like ethical restrictions, high cost, difficult maintenance of large quantity and lesser reproducibility. Besides, various lower chordate animals like Danio rerio, Drosophila melanogaster, Caenorhabditis elegans and Ciona intestinalis have been proved to be an important model for the in vivo determination of targets of drugs with least limitations. In this article, we reviewed different studies conducted on theses models for the better understanding of the pathophysiology of AD and their subsequent application as a potential tool in the preclinical evaluation of new drugs.
Collapse
Affiliation(s)
- Neha Sharma
- Department of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Navneet Khurana
- Department of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Arunachalam Muthuraman
- Department of Pharmacology, Akal College of Pharmacy and Technical Education, Mastuana Sahib, Sangrur, Punjab, India; Department of Pharmacology, JSS College of Pharmacy, Jagadguru Sri Shivarathreeshwara University, Mysuru 570015, Karnataka, India.
| |
Collapse
|
77
|
Decreased gene expression of CD2AP in Chinese patients with sporadic Alzheimer's disease. Neurobiol Aging 2017; 56:212.e5-212.e10. [DOI: 10.1016/j.neurobiolaging.2017.03.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/23/2017] [Accepted: 03/08/2017] [Indexed: 01/07/2023]
|
78
|
Monsell SE, Mock C, Fardo DW, Bertelsen S, Cairns NJ, Roe CM, Ellingson SR, Morris JC, Goate AM, Kukull WA. Genetic Comparison of Symptomatic and Asymptomatic Persons With Alzheimer Disease Neuropathology. Alzheimer Dis Assoc Disord 2017; 31:232-238. [PMID: 27849641 PMCID: PMC5432419 DOI: 10.1097/wad.0000000000000179] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The objective was to determine whether symptomatic and asymptomatic persons with Alzheimer disease (AD) neuropathology have different allele counts for single-nucleotide polymorphisms that have been associated with clinical late-onset AD. METHODS Data came from the National Alzheimer's Coordinating Center Uniform Data Set and Neuropathology Data Set, and the Alzheimer's Disease Genetics Consortium (ADGC). Participants had low to high AD neuropathologic change. The 22 known/suspected genes associated with late-onset AD were considered. "Symptomatic" was defined as Clinical Dementia Rating global score >0. RESULTS Sixty-eight asymptomatic and 521 symptomatic participants met inclusion criteria. Single-nucleotide polymorphisms associated with ABCA7 [odds ratio (OR)=1.66; 95% confidence interval (CI), 1.03-2.85] and MAPT (OR=2.18; CI, 1.26-3.77) were associated with symptomatic status. In stratified analyses, loci containing CD2AP (OR=0.35; 95% CI, 0.16-0.74), ZCWPW1 (OR=2.98; 95% CI, 1.34-6.86), and MAPT (OR=3.73, 95% CI, 1.30-11.76) were associated with symptomatic status in APOE e4 carriers. CONCLUSIONS These findings potentially explain some of the variation in whether a person with AD neuropathology expresses symptoms. Understanding why some people remain cognitively normal despite having AD neuropathology could identify pathways to disease heterogeneity and guide treatment trials.
Collapse
Affiliation(s)
- Sarah E. Monsell
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Charles Mock
- National Alzheimer's Coordinating Center, University of Washington, Seattle, Washington
| | - David W. Fardo
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky
| | - Sarah Bertelsen
- Dept. of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nigel J. Cairns
- Knight Alzheimer's Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Catherine M. Roe
- Knight Alzheimer's Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Sally R. Ellingson
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky
| | - John C. Morris
- Knight Alzheimer's Disease Research Center, Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Alison M. Goate
- Dept. of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Walter A. Kukull
- National Alzheimer's Coordinating Center, University of Washington, Seattle, Washington
| |
Collapse
|
79
|
Perez SE, Nadeem M, He B, Miguel JC, Malek-Ahmadi MH, Chen K, Mufson EJ. Neocortical and hippocampal TREM2 protein levels during the progression of Alzheimer's disease. Neurobiol Aging 2017; 54:133-143. [PMID: 28365005 PMCID: PMC6344038 DOI: 10.1016/j.neurobiolaging.2017.02.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/14/2017] [Accepted: 02/16/2017] [Indexed: 01/09/2023]
Abstract
Heterozygous triggering receptor expressed on myeloid cells (TREM2) mutations are an Alzheimer's disease (AD) risk factor. Nonmutated TREM2 dysregulation occurs in AD brain. Whether TREM2 is altered in prodromal AD remains unknown. Western blotting was used to determine levels of TREM2 (∼25 kDa) and Iba1 in the frontal cortex and TREM2 in the hippocampus from people who died with an ante-mortem clinical diagnosis of non- and mild-cognitive impairment, mild/moderate AD, and severe AD (sAD). Immunohistochemistry defined the relationship between amyloid and Iba1 profiles. Polymerase chain reaction analysis revealed that all subjects did not carry the most common R47H TREM2 variant. TREM2 was significantly upregulated in sAD frontal cortex but stable in hippocampus. Frontal TREM2 mRNA and protein level patterns were similar but not significantly different. Iba1 immunopositive microglia counts increased significantly in frontal cortex containing plaques in sAD. TREM2 and Iba1 levels were not associated with plaques, tangles, neuropathological criteria, or cognitive performance. Frontal cortex TREM2 upregulation is a late event and may not play a major role early in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Sylvia E Perez
- Departments of Neurobiology and Neurology, Alzheimer's disease Research Laboratory, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Muhammad Nadeem
- Departments of Neurobiology and Neurology, Alzheimer's disease Research Laboratory, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Bin He
- Departments of Neurobiology and Neurology, Alzheimer's disease Research Laboratory, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Jennifer C Miguel
- Departments of Neurobiology and Neurology, Alzheimer's disease Research Laboratory, Barrow Neurological Institute, Phoenix, AZ, USA
| | | | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Elliott J Mufson
- Departments of Neurobiology and Neurology, Alzheimer's disease Research Laboratory, Barrow Neurological Institute, Phoenix, AZ, USA.
| |
Collapse
|
80
|
Hinz FI, Geschwind DH. Molecular Genetics of Neurodegenerative Dementias. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a023705. [PMID: 27940516 DOI: 10.1101/cshperspect.a023705] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurodegenerative dementias are clinically heterogeneous, progressive diseases with frequently overlapping symptoms, such as cognitive impairments and behavior and movement deficits. Although a majority of cases appear to be sporadic, there is a large genetic component that has yet to be fully explained. Here, we review the recent genetic and genomic findings pertaining to Alzheimer's disease, frontotemporal dementia, Lewy body dementia, and prion dementia. In this review, we describe causal and susceptibility genes identified for these dementias and discuss recent research pertaining to the molecular function of these genes. Of particular interest, there is a large overlap in clinical phenotypes, genes, and/or aggregating protein products involved in these diseases, as well as frequent comorbid presentation, indicating that these dementias may represent a continuum of syndromes rather than individual diseases.
Collapse
Affiliation(s)
- Flora I Hinz
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095.,Center for Autism Research and Treatment and Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California 90024
| |
Collapse
|
81
|
Šimić G, Babić Leko M, Wray S, Harrington CR, Delalle I, Jovanov-Milošević N, Bažadona D, Buée L, de Silva R, Di Giovanni G, Wischik CM, Hof PR. Monoaminergic neuropathology in Alzheimer's disease. Prog Neurobiol 2017; 151:101-138. [PMID: 27084356 PMCID: PMC5061605 DOI: 10.1016/j.pneurobio.2016.04.001] [Citation(s) in RCA: 201] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 03/09/2016] [Accepted: 04/05/2016] [Indexed: 01/02/2023]
Abstract
None of the proposed mechanisms of Alzheimer's disease (AD) fully explains the distribution patterns of the neuropathological changes at the cellular and regional levels, and their clinical correlates. One aspect of this problem lies in the complex genetic, epigenetic, and environmental landscape of AD: early-onset AD is often familial with autosomal dominant inheritance, while the vast majority of AD cases are late-onset, with the ε4 variant of the gene encoding apolipoprotein E (APOE) known to confer a 5-20 fold increased risk with partial penetrance. Mechanisms by which genetic variants and environmental factors influence the development of AD pathological changes, especially neurofibrillary degeneration, are not yet known. Here we review current knowledge of the involvement of the monoaminergic systems in AD. The changes in the serotonergic, noradrenergic, dopaminergic, histaminergic, and melatonergic systems in AD are briefly described. We also summarize the possibilities for monoamine-based treatment in AD. Besides neuropathologic AD criteria that include the noradrenergic locus coeruleus (LC), special emphasis is given to the serotonergic dorsal raphe nucleus (DRN). Both of these brainstem nuclei are among the first to be affected by tau protein abnormalities in the course of sporadic AD, causing behavioral and cognitive symptoms of variable severity. The possibility that most of the tangle-bearing neurons of the LC and DRN may release amyloid β as well as soluble monomeric or oligomeric tau protein trans-synaptically by their diffuse projections to the cerebral cortex emphasizes their selective vulnerability and warrants further investigations of the monoaminergic systems in AD.
Collapse
Affiliation(s)
- Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.
| | - Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Selina Wray
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | | | - Ivana Delalle
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Nataša Jovanov-Milošević
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Danira Bažadona
- Department of Neurology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Luc Buée
- University of Lille, Inserm, CHU-Lille, UMR-S 1172, Alzheimer & Tauopathies, Lille, France
| | - Rohan de Silva
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Giuseppe Di Giovanni
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Claude M Wischik
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Patrick R Hof
- Fishberg Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
82
|
Mukherjee S, Russell JC, Carr DT, Burgess JD, Allen M, Serie DJ, Boehme KL, Kauwe JSK, Naj AC, Fardo DW, Dickson DW, Montine TJ, Ertekin-Taner N, Kaeberlein MR, Crane PK. Systems biology approach to late-onset Alzheimer's disease genome-wide association study identifies novel candidate genes validated using brain expression data and Caenorhabditis elegans experiments. Alzheimers Dement 2017; 13:1133-1142. [PMID: 28242297 DOI: 10.1016/j.jalz.2017.01.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/27/2016] [Accepted: 01/12/2017] [Indexed: 01/08/2023]
Abstract
INTRODUCTION We sought to determine whether a systems biology approach may identify novel late-onset Alzheimer's disease (LOAD) loci. METHODS We performed gene-wide association analyses and integrated results with human protein-protein interaction data using network analyses. We performed functional validation on novel genes using a transgenic Caenorhabditis elegans Aβ proteotoxicity model and evaluated novel genes using brain expression data from people with LOAD and other neurodegenerative conditions. RESULTS We identified 13 novel candidate LOAD genes outside chromosome 19. Of those, RNA interference knockdowns of the C. elegans orthologs of UBC, NDUFS3, EGR1, and ATP5H were associated with Aβ toxicity, and NDUFS3, SLC25A11, ATP5H, and APP were differentially expressed in the temporal cortex. DISCUSSION Network analyses identified novel LOAD candidate genes. We demonstrated a functional role for four of these in a C. elegans model and found enrichment of differentially expressed genes in the temporal cortex.
Collapse
Affiliation(s)
| | - Joshua C Russell
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Daniel T Carr
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Jeremy D Burgess
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Mariet Allen
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Daniel J Serie
- Department of Health Sciences Research, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Kevin L Boehme
- Department of Biology, Brigham Young University, Provo, Utah, USA; Department of Neuroscience, Brigham Young University, Provo, Utah, USA
| | - John S K Kauwe
- Department of Biology, Brigham Young University, Provo, Utah, USA; Department of Neuroscience, Brigham Young University, Provo, Utah, USA
| | - Adam C Naj
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David W Fardo
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Thomas J Montine
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Nilufer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida, USA; Department of Neurology, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Matt R Kaeberlein
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
83
|
Barrett MJ, Koeppel AF, Flanigan JL, Turner SD, Worrall BB. Investigation of Genetic Variants Associated with Alzheimer Disease in Parkinson Disease Cognition. JOURNAL OF PARKINSONS DISEASE 2017; 6:119-24. [PMID: 26889634 DOI: 10.3233/jpd-150706] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Meta-analysis of genome-wide association studies have implicated multiple single nucleotide polymorphisms (SNPs) and associated genes with Alzheimer disease. The role of these SNPs in cognitive impairment in Parkinson disease (PD) remains incompletely evaluated. OBJECTIVE The objective of this study was to test alleles associated with risk of Alzheimer disease for association with cognitive impairment in Parkinson disease (PD). METHODS Two datasets with PD subjects accessed through the NIH database of Genotypes and Phenotypes contained both single nucleotide polymorphism (SNP) arrays and mini-mental state exam (MMSE) scores. Genetic data underwent rigorous quality control and we selected SNPs for genes associated with AD other than APOE. We constructed logistic regression and ordinal regression models, adjusted for sex, age at MMSE, and duration of PD, to assess the association between selected SNPs and MMSE score. RESULTS In one dataset, PICALM rs3851179 was associated with cognitive impairment (MMSE < 24) in PD subjects > 70 years old (OR = 2.3; adjusted p-value = 0.017; n = 250) but not in PD subjects ≤ 70 years old. CONCLUSIONS Our finding suggests that PICALM rs3851179 could contribute to cognitive impairment in older patients with PD. It is important that future studies consider the interaction of age and genetic risk factors in the development of cognitive impairment in PD.
Collapse
Affiliation(s)
- Matthew J Barrett
- Department of Neurology, University of Virginia, Charlottesville, VA, USA
| | - Alexander F Koeppel
- Bioinformatics Core, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Joseph L Flanigan
- Department of Neurology, University of Virginia, Charlottesville, VA, USA
| | - Stephen D Turner
- Bioinformatics Core, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Bradford B Worrall
- Department of Neurology, University of Virginia, Charlottesville, VA, USA.,Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
84
|
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease in the world. The "amyloid hypothesis" is one of the predominant hypotheses for the pathogenesis of AD. Besides, tau protein accumulation, calcium homeostasis disruption, and glial cell activation are also remarkable features in AD. Recently, there are some reports showing that TRPC channels may function in AD development, especially TRPC6. In this chapter, we will discuss the evidence for the involvement of TRPC channels in Alzheimer's disease and the potential of therapeutics for AD based on TRPC channels.
Collapse
|
85
|
Han J, Wang M, Ren M, Lou H. Contributions of triggering-receptor-expressed-on-myeloid-cells-2 to neurological diseases. Int J Neurosci 2016; 127:368-375. [PMID: 27871212 DOI: 10.1080/00207454.2016.1264072] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Jie Han
- Department of Pharmacology, School of Medicine, Shandong University, Jinan 250012, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute affiliated to Shandong University, Jinan 250012, China
| | - Miaomiao Wang
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Manru Ren
- Department of Pharmacology, School of Medicine, Shandong University, Jinan 250012, China
| | - Haiyan Lou
- Department of Pharmacology, School of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
86
|
Role of Plasma Clusterin in Alzheimer's Disease-A Pilot Study in a Tertiary Hospital in Northern India. PLoS One 2016; 11:e0166369. [PMID: 27861589 PMCID: PMC5115728 DOI: 10.1371/journal.pone.0166369] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 10/27/2016] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To evaluate the role of plasma clusterin in Alzheimer's disease (AD). BACKGROUND Plasma clusterin is a promising biomarker as various studies have shown it to be associated with AD. But other studies have shown that plasma clusterin levels were not related to Alzheimer's disease or presymptomatic AD. Hence the diagnostic value of plasma clusterin is still not conclusive. METHODS Neuropsychological assessment, MRI brain, FDG-PET brain and CSF biomarkers of AD were used for establishing the diagnosis of MCI, AD or Vascular dementia. The CSF control group included patients who were having knee or hip surgery and plasma control group included the spouses of patients. RESULTS Forty-six patients who gave consent for CSF examination and FDG PET brain were included in the study along with 19 control samples. Alzheimer's group had 34 patients and Vascular group had 12 patients. Both had a significantly lower value of clusterin than the control samples (p<0.01). The median plasma clusterin level was 84.38 μg/ml in control group, 57.98μg/ml in Alzheimer's group and 49.93μg/ml in the vascular group. Alzheimer and Vascular group did not differ in plasma clusterin levels. Moreover there was no correlation of plasma clusterin with AD severity. The sensitivity and specificity of plasma clusterin was low for any significance for clinical use. CONCLUSION Our pilot study shows that plasma clusterin is lower in Alzheimer's disease with respect to control population. Plasma clusterin levels and severity of Alzheimer's disease had no significant correlation. There was no difference in plasma clusterin between Alzheimer's disease and Vascular Dementia. The sensitivity and specificity of plasma clusterin is low for any use in clinical practice. More studies are required to ascertain the utility of plasma clusterin as a biomarker in Alzheimer's disease.
Collapse
|
87
|
Villamil-Ortiz JG, Barrera-Ocampo A, Piedrahita D, Velásquez-Rodríguez CM, Arias-Londoño JD, Cardona-Gómez GP. BACE1 RNAi Restores the Composition of Phosphatidylethanolamine-Derivates Related to Memory Improvement in Aged 3xTg-AD Mice. Front Cell Neurosci 2016; 10:260. [PMID: 27891075 PMCID: PMC5105502 DOI: 10.3389/fncel.2016.00260] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 10/26/2016] [Indexed: 01/16/2023] Open
Abstract
β-amyloid (Aβ) is produced by the β-secretase 1 (BACE1)-mediated enzymatic cleavage of the amyloid precursor protein through the amyloidogenic pathway, making BACE1 a therapeutic target against Alzheimer’s disease (AD). Alterations in lipid metabolism are a risk factor for AD by an unknown mechanism. The objective of this study was to determine the effect of RNA interference against BACE1 (shBACEmiR) on the phospholipid profile in hippocampal CA1 area in aged 3xTg-AD mice after 6 and 12 months of treatment compared to aged PS1KI mice. The shBACEmiR treatment induced cognitive function recovery and restored mainly the fatty acid composition of lysophosphatidylethanolamine and etherphosphatidylethanolamine, reduced the cPLA2’s phosphorylation, down-regulated the levels of arachidonic acid and COX2 in the hippocampi of 3xTg-AD mice. Together, our findings suggest, for the first time, that BACE1 silencing restores phospholipids composition which could favor the recovery of cellular homeostasis and cognitive function in the hippocampus of triple transgenic AD mice.
Collapse
Affiliation(s)
- Javier G Villamil-Ortiz
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, Sede de Investigación Universitaria, University of Antioquia Medellín, Colombia
| | - Alvaro Barrera-Ocampo
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, Sede de Investigación Universitaria, University of Antioquia Medellín, Colombia
| | - Diego Piedrahita
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, Sede de Investigación Universitaria, University of Antioquia Medellín, Colombia
| | | | | | - Gloria P Cardona-Gómez
- Cellular and Molecular Neurobiology Area, Group of Neuroscience of Antioquia, Sede de Investigación Universitaria, University of Antioquia Medellín, Colombia
| |
Collapse
|
88
|
Thomas MH, Pelleieux S, Vitale N, Olivier JL. Dietary arachidonic acid as a risk factor for age-associated neurodegenerative diseases: Potential mechanisms. Biochimie 2016; 130:168-177. [PMID: 27473185 DOI: 10.1016/j.biochi.2016.07.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 07/24/2016] [Indexed: 11/28/2022]
Abstract
Alzheimer's disease and associated diseases constitute a major public health concern worldwide. Nutrition-based, preventive strategies could possibly be effective in delaying the occurrence of these diseases and lower their prevalence. Arachidonic acid is the second major polyunsaturated fatty acid (PUFA) and several studies support its involvement in Alzheimer's disease. The objective of this review is to examine how dietary arachidonic acid contributes to Alzheimer's disease mechanisms and therefore to its prevention. First, we explore the sources of neuronal arachidonic acid that could potentially originate from either the conversion of linoleic acid, or from dietary sources and transfer across the blood-brain-barrier. In a second part, a brief overview of the role of the two main agents of Alzheimer's disease, tau protein and Aβ peptide is given, followed by the examination of the relationship between arachidonic acid and the disease. Third, the putative mechanisms by which arachidonic acid could influence Alzheimer's disease occurrence and evolution are presented. The conclusion is devoted to what remains to be determined before integrating arachidonic acid in the design of preventive strategies against Alzheimer's disease and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Mélanie H Thomas
- Unité de Recherche Aliment et Fonctionnalité des Produits Animaux (URAFPA), INRA USC 0340, Université de Lorraine, Nancy, France
| | - Sandra Pelleieux
- Unité de Recherche Aliment et Fonctionnalité des Produits Animaux (URAFPA), INRA USC 0340, Université de Lorraine, Nancy, France
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives (INCI), UPR CNRS 3212, Université de Strasbourg, Strasbourg, France
| | - Jean Luc Olivier
- Unité de Recherche Aliment et Fonctionnalité des Produits Animaux (URAFPA), INRA USC 0340, Université de Lorraine, Nancy, France.
| |
Collapse
|
89
|
Yeh FL, Wang Y, Tom I, Gonzalez LC, Sheng M. TREM2 Binds to Apolipoproteins, Including APOE and CLU/APOJ, and Thereby Facilitates Uptake of Amyloid-Beta by Microglia. Neuron 2016; 91:328-40. [DOI: 10.1016/j.neuron.2016.06.015] [Citation(s) in RCA: 466] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 05/14/2016] [Accepted: 06/07/2016] [Indexed: 11/30/2022]
|
90
|
Wood PL, Medicherla S, Sheikh N, Terry B, Phillipps A, Kaye JA, Quinn JF, Woltjer RL. Targeted Lipidomics of Fontal Cortex and Plasma Diacylglycerols (DAG) in Mild Cognitive Impairment and Alzheimer's Disease: Validation of DAG Accumulation Early in the Pathophysiology of Alzheimer's Disease. J Alzheimers Dis 2016; 48:537-46. [PMID: 26402017 DOI: 10.3233/jad-150336] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Previous studies have demonstrated augmented levels of diacylglycerols (DAG) in the frontal cortex and plasma of Alzheimer's disease (AD) patients. We extended these findings from non-targeted lipidomics studies to design a lipidomics platform to interrogate DAGs and monoacylglycerols (MAG) in the frontal cortex and plasma of MCI subjects. Control subjects included both aged normal controls and controls with normal cognition, but AD pathology at autopsy, individuals termed non-demented AD neuropathology. DAGs with saturated, unsaturated, and polyunsaturated fatty acid substituents were found to be elevated in MCI frontal cortex and plasma. Tandem mass spectrometry of the DAGs did not reveal any differences in the distributions of the fatty acid substitutions between MCI and control subjects. While triacylglycerols were not altered in MCI subjects there were increases in MAG levels both in the frontal cortex and plasma. In toto, increased levels of DAGs and MAGs appear to occur early in AD pathophysiology and require both further validation in a larger patient cohort and elucidation of the lipidomics alteration(s) that lead to the accumulation of DAGs in MCI subjects.
Collapse
Affiliation(s)
- Paul L Wood
- Lipidomics Unit, Department of Physiology and Pharmacology, DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN, USA
| | - Srikanth Medicherla
- Lipidomics Unit, Department of Physiology and Pharmacology, DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN, USA
| | - Naveen Sheikh
- Lipidomics Unit, Department of Physiology and Pharmacology, DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN, USA
| | - Bradley Terry
- Lipidomics Unit, Department of Physiology and Pharmacology, DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN, USA
| | - Aaron Phillipps
- Lipidomics Unit, Department of Physiology and Pharmacology, DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN, USA
| | - Jeffrey A Kaye
- Department of Neurology, Oregon Health Science University and Portland VA Medical Center, Portland, OR, USA
| | - Joseph F Quinn
- Department of Neurology, Oregon Health Science University and Portland VA Medical Center, Portland, OR, USA
| | - Randall L Woltjer
- Department of Neurology, Oregon Health Science University and Portland VA Medical Center, Portland, OR, USA
| |
Collapse
|
91
|
Saadat M. Distributions of susceptibility loci to late onset Alzheimer's disease on human chromosomes. EXCLI JOURNAL 2016; 15:403-5. [PMID: 27540352 PMCID: PMC4983801 DOI: 10.17179/excli2016-161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 06/21/2016] [Indexed: 02/05/2023]
Affiliation(s)
- Mostafa Saadat
- Department of Biology, College of Sciences, Shiraz University, Shiraz 71467-13565, Iran
| |
Collapse
|
92
|
Developmental exposure to lead (Pb) alters the expression of the human tau gene and its products in a transgenic animal model. Neurotoxicology 2016; 55:154-159. [PMID: 27293183 DOI: 10.1016/j.neuro.2016.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/11/2016] [Accepted: 06/01/2016] [Indexed: 10/21/2022]
Abstract
Tauopathies are a class of neurodegenerative diseases associated with the pathological aggregation of the tau protein in the human brain. The best known of these illnesses is Alzheimer's disease (AD); a disease where the microtubule associated protein tau (MAPT) becomes hyperphosphorylated (lowering its binding affinity to microtubules) and aggregates within neurons in the form of neurofibrillary tangles (NFTs). In this paper we examine whether environmental factors play a significant role in tau pathogenesis. Our studies were conducted in a double mutant mouse model that expressed the human tau gene and lacked the gene for murine tau. The human tau mouse model was tested for the transgene's ability to respond to an environmental toxicant. Pups were developmentally exposed to lead (Pb) from postnatal day (PND) 1-20 with 0.2% Pb acetate. Mice were then sacrificed at PND 20, 30, 40 and 60. Protein and mRNA levels for tau and CDK5 as well as tau phosphorylation at Ser396 were determined. In addition, the potential role of miRNA in tau expression was investigated by measuring levels of miR-34c, a miRNA that targets the mRNA for human tau, at PND20 and 50. The expression of the human tau transgene was altered by developmental exposure to Pb. This exposure also altered the expression of miR-34c. Our findings are the first of their kind to test the responsiveness of the human tau gene to an environmental toxicant and to examine an epigenetic mechanism that may be involved in the regulation of this gene's expression.
Collapse
|
93
|
Freudenberg-Hua Y, Li W, Abhyankar A, Vacic V, Cortes V, Ben-Avraham D, Koppel J, Greenwald B, Germer S, Darnell RB, Barzilai N, Freudenberg J, Atzmon G, Davies P. Differential burden of rare protein truncating variants in Alzheimer's disease patients compared to centenarians. Hum Mol Genet 2016; 25:3096-3105. [PMID: 27260402 DOI: 10.1093/hmg/ddw150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 05/06/2016] [Accepted: 05/13/2016] [Indexed: 12/31/2022] Open
Abstract
We compared coding region variants of 53 cognitively healthy centenarians and 45 patients with Alzheimer's disease (AD), all of Ashkenazi Jewish (AJ) ancestry. Despite the small sample size, the known AD risk variant APOE4 reached genome-wide significance, indicating the advantage of utilizing 'super-controls'. We restricted our subsequent analysis to rare variants observed at most once in the 1000 Genomes database and having a minor allele frequency below 2% in our AJ sample. We compared the burden of predicted protein altering variants between cases and controls as normalized by the level of rare synonymous variants. We observed an increased burden among AD subjects for predicted loss-of-function (LoFs) variants defined as stop-gain, frame shift, initiation codon (INIT) and splice site mutations (n = 930, OR = 1.3, P = 1.5×E-5). There was no enrichment across all rare protein altering variants defined as missense plus LoFs, in frame indels and stop-loss variants (n = 13 014, OR = 0.97, P = 0.47). Among LoFs, the strongest burden was observed for INIT (OR = 2.16, P = 0.0097) and premature stop variants predicted to cause non-sense-mediated decay in the majority of transcripts (NMD) (OR = 1.98, P = 0.02). Notably, this increased burden of NMD, INIT and splice variants was more pronounced in a set of 1397 innate immune genes (OR = 4.55, P = 0.0043). Further comparison to additional exomes indicates that the difference in LoF burden originated both from the AD and centenarian sample. In summary, we observed an overall increased burden of rare LoFs in AD subjects as compared to centenarians, and this enrichment is more pronounced for innate immune genes.
Collapse
Affiliation(s)
- Yun Freudenberg-Hua
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA .,Division of Geriatric Psychiatry, Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, USA
| | - Wentian Li
- Robert S Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | | | | | - Vanessa Cortes
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Danny Ben-Avraham
- Institute for Aging Research and the Diabetes Research Center, Department of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jeremy Koppel
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Blaine Greenwald
- Division of Geriatric Psychiatry, Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, USA
| | | | | | | | - Nir Barzilai
- Institute for Aging Research and the Diabetes Research Center, Department of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jan Freudenberg
- Robert S Boas Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA.,The Regeneron Genetics Center, Tarrytown, NY 10591, USA and
| | - Gil Atzmon
- Institute for Aging Research and the Diabetes Research Center, Department of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Faculty of Natural Sciences, University of Haifa, Haifa 31905, Israel
| | - Peter Davies
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| |
Collapse
|
94
|
Masoud AM, Bihaqi SW, Machan JT, Zawia NH, Renehan WE. Early-Life Exposure to Lead (Pb) Alters the Expression of microRNA that Target Proteins Associated with Alzheimer’s Disease. J Alzheimers Dis 2016; 51:1257-64. [DOI: 10.3233/jad-151018] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Anwar M. Masoud
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
- Biochemical Technology Program, Faculty of Applied Science, Thamar University, Thamar, Yemen
| | - Syed W. Bihaqi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail, Kingdom of Saudi Arabia
| | - Jason T. Machan
- Lifespan Biostatistics Core and Departments of Orthopaedics and Surgery, Warren Alpert Medical School, Brown University, Providence RI, USA
| | - Nasser H. Zawia
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - William E. Renehan
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| |
Collapse
|
95
|
Van den Bossche T, Sleegers K, Cuyvers E, Engelborghs S, Sieben A, De Roeck A, Van Cauwenberghe C, Vermeulen S, Van den Broeck M, Laureys A, Peeters K, Mattheijssens M, Vandenbulcke M, Vandenberghe R, Martin JJ, De Deyn PP, Cras P, Van Broeckhoven C. Phenotypic characteristics of Alzheimer patients carrying an ABCA7 mutation. Neurology 2016; 86:2126-33. [PMID: 27037232 PMCID: PMC4917260 DOI: 10.1212/wnl.0000000000002628] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 01/12/2016] [Indexed: 11/21/2022] Open
Abstract
Objective: To generate a clinical and pathologic phenotype of patients carrying rare loss-of-function mutations in ABCA7, identified in a Belgian Alzheimer patient cohort and in an autosomal dominant family. Methods: We performed a retrospective review of available data records, medical records, results of CSF analyses and neuroimaging studies, and neuropathology data. Results: The mean onset age of the mutation carriers (n = 22) was 73.4 ± 8.4 years with a wide age range of 36 (54–90) years, which was independent of APOE genotype and cerebrovascular disease. The mean disease duration was 5.7 ± 3.0 years (range 2–12 years). A positive family history was recorded for 10 carriers (45.5%). All patient carriers except one presented with memory complaints. The 4 autopsied brains showed typical immunohistochemical changes of late-onset Alzheimer disease. Conclusions: All patients carrying a loss-of-function mutation in ABCA7 exhibited a classical Alzheimer disease phenotype, though with a striking wide onset age range, suggesting the influence of unknown modifying factors.
Collapse
Affiliation(s)
- Tobi Van den Bossche
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Kristel Sleegers
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Elise Cuyvers
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Sebastiaan Engelborghs
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Anne Sieben
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Arne De Roeck
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Caroline Van Cauwenberghe
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Steven Vermeulen
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Marleen Van den Broeck
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Annelies Laureys
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Karin Peeters
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Maria Mattheijssens
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Mathieu Vandenbulcke
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Rik Vandenberghe
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Jean-Jacques Martin
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Peter P De Deyn
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | - Patrick Cras
- From the Neurodegenerative Brain Diseases Group (T.V.d.B., K.S., E.C., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., C.V.B.), Department of Molecular Genetics, VIB, Antwerp; Institute Born-Bunge (T.V.d.B., K.S., E.C., S.E., A.S., A.D.R., C.V.C., S.V., M.V.d.B., A.L., K.P., M.M., J.-J.M., P.P.D.D., P.C., C.V.B.), University of Antwerp; Department of Neurology (T.V.d.B., P.C.), Antwerp University Hospital, Edegem; Department of Neurology and Memory Clinic (T.V.d.B., S.E., P.P.D.D.), Hospital Netwerk Antwerp (ZNA), Middelheim and Hoge Beuken; Department of Neurology (A.S.), University Hospital Ghent and University of Ghent; Department of Neurosciences (M.V., R.V.), Faculty of Medicine, KU Leuven; Department of Old Age Psychiatry and Memory Clinic (M.V.) and Department of Neurology (R.V.), University Hospitals Leuven, Belgium; and Department of Neurology and Alzheimer Research Center (P.P.D.D.), University of Groningen and University Medical Center Groningen, the Netherlands
| | | | | |
Collapse
|
96
|
Schott JM, Crutch SJ, Carrasquillo MM, Uphill J, Shakespeare TJ, Ryan NS, Yong KX, Lehmann M, Ertekin-Taner N, Graff-Radford NR, Boeve BF, Murray ME, Khan QUA, Petersen RC, Dickson DW, Knopman DS, Rabinovici GD, Miller BL, González AS, Gil-Néciga E, Snowden JS, Harris J, Pickering-Brown SM, Louwersheimer E, van der Flier WM, Scheltens P, Pijnenburg YA, Galasko D, Sarazin M, Dubois B, Magnin E, Galimberti D, Scarpini E, Cappa SF, Hodges JR, Halliday GM, Bartley L, Carrillo MC, Bras JT, Hardy J, Rossor MN, Collinge J, Fox NC, Mead S. Genetic risk factors for the posterior cortical atrophy variant of Alzheimer's disease. Alzheimers Dement 2016; 12:862-71. [PMID: 26993346 PMCID: PMC4982482 DOI: 10.1016/j.jalz.2016.01.010] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 01/26/2016] [Accepted: 01/28/2016] [Indexed: 11/15/2022]
Abstract
Introduction The genetics underlying posterior cortical atrophy (PCA), typically a rare variant of Alzheimer's disease (AD), remain uncertain. Methods We genotyped 302 PCA patients from 11 centers, calculated risk at 24 loci for AD/DLB and performed an exploratory genome-wide association study. Results We confirm that variation in/near APOE/TOMM40 (P = 6 × 10−14) alters PCA risk, but with smaller effect than for typical AD (PCA: odds ratio [OR] = 2.03, typical AD: OR = 2.83, P = .0007). We found evidence for risk in/near CR1 (P = 7 × 10−4), ABCA7 (P = .02) and BIN1 (P = .04). ORs at variants near INPP5D and NME8 did not overlap between PCA and typical AD. Exploratory genome-wide association studies confirmed APOE and identified three novel loci: rs76854344 near CNTNAP5 (P = 8 × 10−10 OR = 1.9 [1.5–2.3]); rs72907046 near FAM46A (P = 1 × 10−9 OR = 3.2 [2.1–4.9]); and rs2525776 near SEMA3C (P = 1 × 10−8, OR = 3.3 [2.1–5.1]). Discussion We provide evidence for genetic risk factors specifically related to PCA. We identify three candidate loci that, if replicated, may provide insights into selective vulnerability and phenotypic diversity in AD.
Collapse
Affiliation(s)
- Jonathan M Schott
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, London, UK.
| | - Sebastian J Crutch
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, London, UK
| | | | - James Uphill
- Department of Neurodegenerative Disease, MRC Prion Unit, UCL Institute of Neurology, London, UK
| | - Tim J Shakespeare
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, London, UK
| | - Natalie S Ryan
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, London, UK
| | - Keir X Yong
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, London, UK
| | - Manja Lehmann
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, London, UK
| | - Nilufer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA; Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | | | | | | | | | | | | | | | | | | | - Aida Suárez González
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, London, UK; Memory Disorders Unit, Department of Neurology, University Hospital Virgen del Rocio, Seville, Spain
| | - Eulogio Gil-Néciga
- Memory Disorders Unit, Department of Neurology, University Hospital Virgen del Rocio, Seville, Spain
| | - Julie S Snowden
- Institute of Brain, Behaviour and Mental Health, University of Manchester, UK
| | - Jenny Harris
- Institute of Brain, Behaviour and Mental Health, University of Manchester, UK
| | | | - Eva Louwersheimer
- Alzheimer Center, Department of Neurology, VU University Medical Center, Neuroscience Campus, Amsterdam, Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center, Department of Neurology, VU University Medical Center, Neuroscience Campus, Amsterdam, Netherlands
| | - Philip Scheltens
- Alzheimer Center, Department of Neurology, VU University Medical Center, Neuroscience Campus, Amsterdam, Netherlands
| | - Yolande A Pijnenburg
- Alzheimer Center, Department of Neurology, VU University Medical Center, Neuroscience Campus, Amsterdam, Netherlands
| | - Douglas Galasko
- Department of Epidemiology & Biostatistics, VU University Medical Center, Amsterdam, The Netherlands; UC San Diego/VA San Diego Healthcare System, San Diego, CA, USA
| | - Marie Sarazin
- INSERM U610, Hôpital de la Salpêtrière, Paris, France
| | - Bruno Dubois
- Centre des Maladies Cognitives et Comportementales, IM2A, ICM, Paris 6 University, France
| | - Eloi Magnin
- Regional Memory Centre (CMRR), CHU Besançon, Besançon, France
| | - Daniela Galimberti
- University of Milan, Fondazione Cà Granda, IRCCS Ospedale Policlinico, Italy
| | - Elio Scarpini
- University of Milan, Fondazione Cà Granda, IRCCS Ospedale Policlinico, Italy
| | | | | | | | | | | | - Jose T Bras
- Department of Molecular Neurosciences, UCL Institute of Neurology, London, UK
| | - John Hardy
- Department of Molecular Neurosciences, UCL Institute of Neurology, London, UK
| | - Martin N Rossor
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, London, UK
| | - John Collinge
- Department of Neurodegenerative Disease, MRC Prion Unit, UCL Institute of Neurology, London, UK
| | - Nick C Fox
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, London, UK
| | - Simon Mead
- Department of Neurodegenerative Disease, MRC Prion Unit, UCL Institute of Neurology, London, UK
| |
Collapse
|
97
|
Piccoli E, Rossi G, Rossi T, Pelliccioni G, D'Amato I, Tagliavini F, Di Fede G. Novel PSEN1 mutations (H214N and R220P) associated with familial Alzheimer's disease identified by targeted exome sequencing. Neurobiol Aging 2016; 40:192.e7-192.e11. [PMID: 26925509 DOI: 10.1016/j.neurobiolaging.2016.01.134] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/30/2015] [Accepted: 01/30/2016] [Indexed: 01/21/2023]
Abstract
Autosomal dominant Alzheimer's disease (AD) is caused by mutations in amyloid precursor protein, presenilin 1 (PSEN1), and presenilin 2 genes and is mostly associated with early-onset form of AD (EOAD), whereas very few mutations were also found in late-onset AD (LOAD) cases. Because of the clinical overlapping between AD and other degenerative dementias such as frontotemporal dementias, a wide-spectrum genetic analysis should be envisaged in the differential diagnosis of this group of disorders. We used next-generation sequencing techniques to analyze 10 genes involved in dementia on a cohort of 20 EOAD and 20 LOAD cases. We found 5 rare coding variants (frequency <1%). PSEN1 H214N mutation, identified in a case of familial EOAD and PSEN1 R220P, found in a case of familial LOAD, are predicted to be pathogenic. These findings confirm the contribution of PSEN1 genetic variants also to LOAD, underlining the need of extending the genetic screening of presenilin mutations to LOAD patients. Two variants in microtubule-associated protein tau and 1 in progranulin appeared to be benign polymorphisms, showing no major contribution of these genes to AD.
Collapse
Affiliation(s)
- Elena Piccoli
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Giacomina Rossi
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy.
| | - Tommaso Rossi
- Division of Neurology, Geriatric Hospital, INRCA IRCCS, Ancona, Italy
| | | | - Ilaria D'Amato
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Fabrizio Tagliavini
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Giuseppe Di Fede
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| |
Collapse
|
98
|
Eid A, Bihaqi SW, Renehan WE, Zawia NH. Developmental lead exposure and lifespan alterations in epigenetic regulators and their correspondence to biomarkers of Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2016; 2:123-31. [PMID: 27239543 PMCID: PMC4879653 DOI: 10.1016/j.dadm.2016.02.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
INTRODUCTION Early life lead (Pb) exposure results in a latent increase in Alzheimer's disease (AD)-related proteins, and cognitive deficits late in life in both rodents and primates. This study was conducted to investigate if these late life changes were accompanied by epigenetic alterations. METHODS Western blot analysis and RT-PCR were used to measure Deoxyribonucleic acid methylation regulators (DNMT1, DNMT3a, MeCP2, MAT2A) and histone proteins (H3K9Ac, H3K4me2, H3K27me3). RESULTS Cerebral levels of DNMT1 and MeCP2 were significantly reduced in mice exposed to Pb early in life, whereas the expression of DNMT3a was not altered. Levels of MAT2a were increased in the Pb-exposed mice across the lifespan. H3K9Ac and H3K4me2, involved in gene activation, were decreased, whereas the repressive mark H3K27me3 was elevated. DISCUSSION Epigenetic modifiers are affected by the developmental exposure to Pb and may play a role in mediating the latent increases in AD-related proteins in the brain.
Collapse
Affiliation(s)
- Aseel Eid
- Neurodegeneration Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
- Geroge and Ann Ryan Institute for Neuroscience, University of Rhode Island, Kingston RI, USA
| | - Syed Waseem Bihaqi
- Department of Pharmacology and Toxicology, University of Hail, Hail, Kingdom of Saudi Arabia
| | - William E. Renehan
- Neurodegeneration Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
- Geroge and Ann Ryan Institute for Neuroscience, University of Rhode Island, Kingston RI, USA
| | - Nasser H. Zawia
- Neurodegeneration Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
- Geroge and Ann Ryan Institute for Neuroscience, University of Rhode Island, Kingston RI, USA
| |
Collapse
|
99
|
Nagamine S, Yamazaki T, Makioka K, Fujita Y, Ikeda M, Takatama M, Okamoto K, Yokoo H, Ikeda Y. Hypersialylation is a common feature of neurofibrillary tangles and granulovacuolar degenerations in Alzheimer's disease and tauopathy brains. Neuropathology 2015; 36:333-45. [DOI: 10.1111/neup.12277] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 10/20/2015] [Accepted: 10/20/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Shun Nagamine
- Department of Neurology; Gunma University Graduate School of Medicine; Maebashi Gunma Japan
| | - Tsuneo Yamazaki
- Department of Rehabilitation; Gunma University Graduate School of Health Sciences; Maebashi Gunma Japan
| | - Kouki Makioka
- Department of Neurology; Gunma University Graduate School of Medicine; Maebashi Gunma Japan
| | - Yukio Fujita
- Department of Neurology; Gunma University Graduate School of Medicine; Maebashi Gunma Japan
| | - Masaki Ikeda
- Department of Neurology; Gunma University Graduate School of Medicine; Maebashi Gunma Japan
| | - Masamitsu Takatama
- Geriatrics Research Institute and Hospital; Gunma University Graduate School of Medicine
| | - Koichi Okamoto
- Geriatrics Research Institute and Hospital; Gunma University Graduate School of Medicine
| | - Hideaki Yokoo
- Department of Human Pathology; Gunma University Graduate School of Medicine; Maebashi Gunma Japan
| | - Yoshio Ikeda
- Department of Neurology; Gunma University Graduate School of Medicine; Maebashi Gunma Japan
| |
Collapse
|
100
|
Ramirez LM, Goukasian N, Porat S, Hwang KS, Eastman JA, Hurtz S, Wang B, Vang N, Sears R, Klein E, Coppola G, Apostolova LG. Common variants in ABCA7 and MS4A6A are associated with cortical and hippocampal atrophy. Neurobiol Aging 2015; 39:82-9. [PMID: 26923404 DOI: 10.1016/j.neurobiolaging.2015.10.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 10/01/2015] [Accepted: 10/30/2015] [Indexed: 12/25/2022]
Abstract
The precise physiologic function of many of the recently discovered Alzheimer's disease risk variants remains unknown. The downstream effects of genetic variants remain largely unexplored. We studied the relationship between the top 10 non-APOE genes with cortical and hippocampal atrophy as markers of neurodegeneration using 1.5T magnetic resonance imaging, 1-million single nucleotide polymorphism Illumina Human Omni-Quad array and Illumina Human BeadChip peripheral blood expression array data on 50 cognitively normal and 98 mild cognitive impairment subjects. After explicit matching of cortical and hippocampal morphology, we computed in 3D, the cortical thickness and hippocampal radial distance measures for each participant. Associations between the top 10 non-APOE genome-wide hits and neurodegeneration were explored using linear regression. Map-wise statistical significance was determined with permutations using threshold of p < 0.01. MS4A6A rs610932 and ABCA7 rs3764650 demonstrated significant associations with cortical and hippocampal atrophy. Exploratory MS4A6A and ABCA7 peripheral blood expression analyses revealed a similar pattern of associations with cortical neurodegeneration. To our knowledge, this is the first report of the effect of ABCA7 and MS4A6A on neurodegeneration.
Collapse
Affiliation(s)
| | | | - Shai Porat
- Department of Neurology, UCLA, Los Angeles, CA, USA
| | - Kristy S Hwang
- Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | | | - Sona Hurtz
- Drexel University College of Medicine, Philadelphia, PA, USA
| | | | - Nouchee Vang
- Department of Family Medicine, University of Minnesota, Twin Cities, MN, USA
| | - Renee Sears
- Department of Psychiatry, UCLA, Los Angeles, CA, USA
| | - Eric Klein
- Department of Psychiatry, UCLA, Los Angeles, CA, USA
| | | | - Liana G Apostolova
- Department of Neurology, UCLA, Los Angeles, CA, USA; Department of Neurology, Indiana University, Indianapolis, IN, USA; Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|