51
|
Nettiksimmons J, Tranah G, Evans DS, Yokoyama JS, Yaffe K. Gene-based aggregate SNP associations between candidate AD genes and cognitive decline. AGE (DORDRECHT, NETHERLANDS) 2016; 38:41. [PMID: 27005436 PMCID: PMC5005889 DOI: 10.1007/s11357-016-9885-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/28/2016] [Indexed: 05/08/2023]
Abstract
Single nucleotide polymorphisms (SNPs) in and near ABCA7, BIN1, CASS4, CD2AP, CD33, CELF1, CLU, complement receptor 1 (CR1), EPHA1, EXOC3L2, FERMT2, HLA cluster (DRB5-DQA), INPP5D, MEF2C, MS4A cluster (MS4A3-MS4A6E), NME8, PICALM, PTK2B, SLC24A4, SORL1, and ZCWPW1 have been associated with Alzheimer's disease (AD) in large meta-analyses. We aimed to determine whether established AD-associated genes are associated with longitudinal cognitive decline by examining aggregate variation across these gene regions. In two single-sex cohorts of older, community-dwelling adults, we examined the association between SNPs in previously implicated gene regions and cognitive decline (age-adjusted person-specific cognitive slopes) using a Sequence Kernel Association Test (SKAT). In regions which showed aggregate significance, we examined the univariate association between individual SNPs in the region and cognitive decline. Only two of the original AD-associated SNPs were significantly associated with cognitive decline in our cohorts. We identified significant aggregate-level associations between cognitive decline and the gene regions BIN1, CD33, CELF1, CR1, HLA cluster, and MEF2C in the all-female cohort and significant associations with ABCA7, HLA cluster, MS4A6E, PICALM, PTK2B, SLC24A4, and SORL1 in the all-male cohort. We also identified a block of eight correlated SNPs in CD33 and several blocks of correlated SNPs in CELF1 that were significantly associated with cognitive decline in univariate analysis in the all-female cohort.
Collapse
Affiliation(s)
- Jasmine Nettiksimmons
- Department of Psychiatry, University of San Francisco - California, 4150 Clement Street, Box VAMC-116H, San Francisco, CA 94121 USA
| | - Gregory Tranah
- California Pacific Medical Center Research Institute, Department of Epidemiology and Biostatistics, University of California - San Francisco, Mission Hall: Global Health & Clinical Sciences Building, 550 16th Street, 2nd floor, Box #0560, San Francisco, CA 94158-2549 USA
| | - Daniel S. Evans
- California Pacific Medical Center Research Institute, Mission Hall: Global Health & Clinical Sciences Building, 550 16th Street, 2nd floor, Box #0560, San Francisco, CA 94158-2549 USA
| | - Jennifer S. Yokoyama
- Memory and Aging Center, University of California - San Francisco, Sandler Neurosciences Center, 675 Nelson Rising Lane, Suite 190, San Francisco, CA USA
| | - Kristine Yaffe
- Departments of Psychiatry, Neurology, and Epidemiology and Biostatistics, University of California - San Francisco, San Francisco Veterans Affairs Medical Center, 4150 Clement Street, Box 181, San Francisco, CA 94121 USA
| |
Collapse
|
52
|
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: 9.4] [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
|
53
|
Li JQ, Wang HF, Zhu XC, Sun FR, Tan MS, Tan CC, Jiang T, Tan L, Yu JT. GWAS-Linked Loci and Neuroimaging Measures in Alzheimer's Disease. Mol Neurobiol 2016; 54:146-153. [PMID: 26732597 DOI: 10.1007/s12035-015-9669-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 12/17/2015] [Indexed: 01/01/2023]
Abstract
Recently, 19 susceptibility loci for Alzheimer's disease (AD) had been identified through AD genome-wide association studies (GWAS) meta-analysis. However, how they influence the pathogenesis of AD still remains largely unknown. We studied those loci with six MRI measures, abnormal glucose metabolism, and β-amyloid (Aβ) deposition on neuroimaging in a large cohort from Alzheimer's Disease Neuroimaging Initiative (ADNI) database in order to provide clues of the mechanisms through which these genetic variants might be acting. As a result, single nucleotide polymorphisms (SNPs) at rs983392 within MS4A6A and rs11218343 within SOLR1 were both associated with the percentage of increase in the volume of left inferior temporal regions in the follow-up study. Meanwhile, rs11218343 at SORL1 and rs6733839 at BIN1 was associated with rate of volume change of left parahippocampal and right inferior parietal, respectively. Moreover, rs6656401 at CR1 and rs983392 at MS4A6A were both associated with smaller volume of right middle temporal at baseline. However, in addition to the APOE locus, we did not detect any influence on glucose metabolism and Aβ deposition. APOE ε4 allele was associated with almost all measures. Altogether, five loci (rs6656401 at CR1, rs983392within MS4A6A, rs11218343 at SORL1, rs6733839 at BIN1, and APOE ε4) have been detected to be associated with one or a few established AD-related neuroimaging measures.
Collapse
Affiliation(s)
- Jie-Qiong Li
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, Shandong Province, 266071, China
| | - Hui-Fu Wang
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao, China
| | - Xi-Chen Zhu
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao, China
| | - Fu-Rong Sun
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, Shandong Province, 266071, China
| | - Meng-Shan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, Shandong Province, 266071, China
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, Shandong Province, 266071, China
| | - Teng Jiang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, Shandong Province, 266071, China.
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, Shandong Province, 266071, China.
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, Suite 190, Box 1207, San Francisco, CA, 94158, USA.
| |
Collapse
|
54
|
Hunter S, Martin S, Brayne C. The APP Proteolytic System and Its Interactions with Dynamic Networks in Alzheimer's Disease. Methods Mol Biol 2016; 1303:71-99. [PMID: 26235060 DOI: 10.1007/978-1-4939-2627-5_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Diseases of aging are often complex and multifactorial, involving many genetic and life course modifiers. Systems biology is becoming an essential tool to investigate disease initiation and disease progression. Alzheimer's disease (AD) can be used as a case study to investigate the application of systems biology to complex disease. Here we describe approaches to capturing biological data, representing data in terms of networks and interpreting their meaning in relation to the human population. We highlight issues that remain to be addressed both in terms of modeling disease progression and in relating findings to the current understanding of human disease.
Collapse
Affiliation(s)
- Sally Hunter
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge School of Clinical Medicine, Forvie Site, Cambridge Biomedical Campus, Box 113, Cambridge, CB2 0SP, UK,
| | | | | |
Collapse
|
55
|
Jiao B, Liu X, Zhou L, Wang MH, Zhou Y, Xiao T, Zhang W, Sun R, Waye MMY, Tang B, Shen L. Polygenic Analysis of Late-Onset Alzheimer's Disease from Mainland China. PLoS One 2015; 10:e0144898. [PMID: 26680604 PMCID: PMC4683047 DOI: 10.1371/journal.pone.0144898] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 11/24/2015] [Indexed: 01/14/2023] Open
Abstract
Recently, a number of single nucleotide polymorphisms (SNPs) were identified to be associated with late-onset Alzheimer disease (LOAD) through genome-wide association study data. Identification of SNP-SNP interaction played an important role in better understanding genetic basis of LOAD. In this study, fifty-eight SNPs were screened in a cohort of 229 LOAD cases and 318 controls from mainland China, and their interaction was evaluated by a series of analysis methods. Seven risk SNPs and six protective SNPs were identified to be associated with LOAD. Risk SNPs included rs9331888 (CLU), rs6691117 (CR1), rs4938933 (MS4A), rs9349407 (CD2AP), rs1160985 (TOMM40), rs4945261 (GAB2) and rs5984894 (PCDH11X); Protective SNPs consisted of rs744373 (BIN1), rs1562990 (MS4A), rs597668 (EXOC3L2), rs9271192 (HLA-DRB5/DRB1), rs157581 and rs11556505 (TOMM40). Among positive SNPs presented above, we found the interaction between rs4938933 (risk) and rs1562990 (protective) in MS4A weakened their each effect for LOAD; for three significant SNPs in TOMM40, their cumulative interaction induced the two protective SNPs effects lost and made the risk SNP effect aggravate for LOAD. Finally, we found rs6656401-rs3865444 (CR1-CD33) pairs were significantly associated with decreasing LOAD risk, while rs28834970-rs6656401 (PTK2B-CR1), and rs28834970-rs6656401 (PTK2B-CD33) were associated with increasing LOAD risk. In a word, our study indicates that SNP-SNP interaction existed in the same gene or cross different genes, which could weaken or aggravate their initial single effects for LOAD.
Collapse
Affiliation(s)
- Bin Jiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyan Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Maggie Haitian Wang
- Division of Biostatistics, School of Public Health and Primary Care, the Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | - Yafang Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Tingting Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Weiwei Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Rui Sun
- Division of Biostatistics, School of Public Health and Primary Care, the Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | - Mary Miu Yee Waye
- School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- State Key Laboratory of Medical Genetics, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- State Key Laboratory of Medical Genetics, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| |
Collapse
|
56
|
Villegas-Llerena C, Phillips A, Garcia-Reitboeck P, Hardy J, Pocock JM. Microglial genes regulating neuroinflammation in the progression of Alzheimer's disease. Curr Opin Neurobiol 2015; 36:74-81. [PMID: 26517285 DOI: 10.1016/j.conb.2015.10.004] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/24/2015] [Accepted: 10/07/2015] [Indexed: 01/09/2023]
Abstract
Neuroinflammation is a pathological hallmark of Alzheimer's disease (AD), and microglia, the brain's resident phagocyte, are pivotal for the immune response observed in AD. Microglia act as sentinel and protective cells, but may become inappropriately reactive in AD to drive neuropathology. Recent Genome Wide Association Studies (GWAS) have identified more than 20 gene variants associated with an increased risk of late-onset AD (LOAD), the most prevalent form of AD [1]. The findings strongly implicate genes related to the immune response (CR1, CD33, MS4A, CLU, ABCA7, EPHA1 and HLA-DRB5-HLA-DRB1), endocytosis (BIN1, PICALM, CD2AP, EPHA1 and SORL1) and lipid biology (CLU, ABCA7 and SORL1) [2-8], and many encode proteins which are highly expressed in microglia [1]. Furthermore, recent identification of a low frequency mutation in the gene encoding the triggering receptor expressed in myeloid cells 2 protein (TREM2) confers increased risk of AD in LOAD cohorts with an effect size similar to that for APOE, until recently the only identified genetic risk factor associated with LOAD [9,10(••)] (Figure 1). The present review summarises our current understanding of the probable roles of microglial genes in the regulation of neuroinflammatory processes in AD and their relation to other processes affecting the disease's progression.
Collapse
Affiliation(s)
- Claudio Villegas-Llerena
- Department of Neuroinflammation, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK; Department of Molecular Neuroscience, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK
| | - Alexandra Phillips
- Department of Neuroinflammation, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK
| | - Pablo Garcia-Reitboeck
- Department of Neuroinflammation, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK; Department of Molecular Neuroscience, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK
| | - John Hardy
- Department of Molecular Neuroscience, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK
| | - Jennifer M Pocock
- Department of Neuroinflammation, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK.
| |
Collapse
|
57
|
Allen M, Kachadoorian M, Carrasquillo MM, Karhade A, Manly L, Burgess JD, Wang C, Serie D, Wang X, Siuda J, Zou F, Chai HS, Younkin C, Crook J, Medway C, Nguyen T, Ma L, Malphrus K, Lincoln S, Petersen RC, Graff-Radford NR, Asmann YW, Dickson DW, Younkin SG, Ertekin-Taner N. Late-onset Alzheimer disease risk variants mark brain regulatory loci. NEUROLOGY-GENETICS 2015; 1:e15. [PMID: 27066552 PMCID: PMC4807909 DOI: 10.1212/nxg.0000000000000012] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/08/2015] [Indexed: 11/15/2022]
Abstract
Objective: To investigate the top late-onset Alzheimer disease (LOAD) risk loci detected or confirmed by the International Genomics of Alzheimer's Project for association with brain gene expression levels to identify variants that influence Alzheimer disease (AD) risk through gene expression regulation. Methods: Expression levels from the cerebellum (CER) and temporal cortex (TCX) were obtained using Illumina whole-genome cDNA-mediated annealing, selection, extension, and ligation assay (WG-DASL) for ∼400 autopsied patients (∼200 with AD and ∼200 with non-AD pathologies). We tested 12 significant LOAD genome-wide association study (GWAS) index single nucleotide polymorphisms (SNPs) for cis association with levels of 34 genes within ±100 kb. We also evaluated brain levels of 14 LOAD GWAS candidate genes for association with 1,899 cis-SNPs. Significant associations were validated in a subset of TCX samples using next-generation RNA sequencing (RNAseq). Results: We identified strong associations of brain CR1, HLA-DRB1, and PILRB levels with LOAD GWAS index SNPs. We also detected other strong cis-SNPs for LOAD candidate genes MEF2C, ZCWPW1, and SLC24A4. MEF2C and SLC24A4, but not ZCWPW1 cis-SNPs, also associate with LOAD risk, independent of the index SNPs. The TCX expression associations could be validated with RNAseq for CR1, HLA-DRB1, ZCWPW1, and SLC24A4. Conclusions: Our results suggest that some LOAD GWAS variants mark brain regulatory loci, nominate genes under regulation by LOAD risk variants, and annotate these variants for their brain regulatory effects.
Collapse
Affiliation(s)
- Mariet Allen
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Michaela Kachadoorian
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Minerva M Carrasquillo
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Aditya Karhade
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Lester Manly
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Jeremy D Burgess
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Chen Wang
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Daniel Serie
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Xue Wang
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Joanna Siuda
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Fanggeng Zou
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - High Seng Chai
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Curtis Younkin
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Julia Crook
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Christopher Medway
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Thuy Nguyen
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Li Ma
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Kimberly Malphrus
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Sarah Lincoln
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Ronald C Petersen
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Neill R Graff-Radford
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Yan W Asmann
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Dennis W Dickson
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Steven G Younkin
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience (M.A., M.K., M.M.C., A.K., L. Manly, J.D.B., J.S., F.Z., C.Y., C.M., T.N., L. Ma, K.M., S.L., D.W.D., S.G.Y., N.E.-T.), Department of Neurology (N.R.G.-R., N.E.-T.), and Health Sciences Research (D.S., X.W., J.C., Y.W.A.), Mayo Clinic, Jacksonville, FL; Department of Neurology (R.C.P.) and Health Sciences Research (C.W., H.S.C.), Mayo Clinic, Rochester, MN; and Department of Neurology (J.S.), Medical University of Silesia, Katowice, Poland
| |
Collapse
|
58
|
Van Giau V, An SSA, Bagyinszky E, Kim S. Gene panels and primers for next generation sequencing studies on neurodegenerative disorders. Mol Cell Toxicol 2015. [DOI: 10.1007/s13273-015-0011-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
59
|
Abstract
Alzheimer's disease (AD) represents the main form of dementia, and is a major public health problem. Despite intensive research efforts, current treatments have only marginal symptomatic benefits and there are no effective disease-modifying or preventive interventions. AD has a strong genetic component, so much research in AD has focused on identifying genetic causes and risk factors. This chapter will cover genetic discoveries in AD and their consequences in terms of improved knowledge regarding the disease and the identification of biomarkers and drug targets. First, we will discuss the study of the rare early-onset, autosomal dominant forms of AD that led to the discovery of mutations in three major genes, APP, PSEN1, and PSEN2. These discoveries have shaped our current understanding of the pathophysiology and natural history of AD as well as the development of therapeutic targets and the design of clinical trials. Then, we will explore linkage analysis and candidate gene approaches, which identified variants in Apolipoprotein E (APOE) as the major genetic risk factor for late-onset, "sporadic" forms of AD (LOAD), but failed to robustly identify other genetic risk factors, with the exception of variants in SORL1. The main focus of this chapter will be on recent genome-wide association studies that have successfully identified common genetic variations at over 20 loci associated with LOAD outside of the APOE locus. These loci are in or near-novel AD genes including BIN1, CR1, CLU, phosphatidylinositol-binding clathrin assembly protein (PICALM), CD33, EPHA1, MS4A4/MS4A6, ABCA7, CD2AP, SORL1, HLA-DRB5/DRB1, PTK2B, SLC24A4-RIN3, INPP5D, MEF2C, NME8, ZCWPW1, CELF1, FERMT2, CASS4, and TRIP4 and each has small effects on risk of AD (relative risks of 1.1-1.3). Finally, we will touch upon the ongoing effort to identify less frequent and rare variants through whole exome and whole genome sequencing. This effort has identified two novel genes, TREM2 and PLD3, and shown a role for APP in LOAD. The identification of these recently identified genes has implicated previously unsuspected biological pathways in the pathophysiology of AD.
Collapse
Affiliation(s)
- Vincent Chouraki
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Framingham Heart Study, Framingham, MA, USA
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Framingham Heart Study, Framingham, MA, USA
| |
Collapse
|
60
|
Wes PD, Holtman IR, Boddeke EW, Möller T, Eggen BJ. Next generation transcriptomics and genomics elucidate biological complexity of microglia in health and disease. Glia 2015; 64:197-213. [DOI: 10.1002/glia.22866] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 05/11/2015] [Indexed: 12/11/2022]
Affiliation(s)
| | - Inge R. Holtman
- Department of NeuroscienceSection Medical Physiology, University of Groningen, University Medical Center GroningenGroningen The Netherlands
| | - Erik W.G.M. Boddeke
- Department of NeuroscienceSection Medical Physiology, University of Groningen, University Medical Center GroningenGroningen The Netherlands
| | | | - Bart J.L. Eggen
- Department of NeuroscienceSection Medical Physiology, University of Groningen, University Medical Center GroningenGroningen The Netherlands
| |
Collapse
|
61
|
Tai LM, Ghura S, Koster KP, Liakaite V, Maienschein‐Cline M, Kanabar P, Collins N, Ben‐Aissa M, Lei AZ, Bahroos N, Green SJ, Hendrickson B, Van Eldik LJ, LaDu MJ. APOE-modulated Aβ-induced neuroinflammation in Alzheimer's disease: current landscape, novel data, and future perspective. J Neurochem 2015; 133:465-88. [PMID: 25689586 PMCID: PMC4400246 DOI: 10.1111/jnc.13072] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 01/12/2023]
Abstract
Chronic glial activation and neuroinflammation induced by the amyloid-β peptide (Aβ) contribute to Alzheimer's disease (AD) pathology. APOE4 is the greatest AD-genetic risk factor; increasing risk up to 12-fold compared to APOE3, with APOE4-specific neuroinflammation an important component of this risk. This editorial review discusses the role of APOE in inflammation and AD, via a literature review, presentation of novel data on Aβ-induced neuroinflammation, and discussion of future research directions. The complexity of chronic neuroinflammation, including multiple detrimental and beneficial effects occurring in a temporal and cell-specific manner, has resulted in conflicting functional data for virtually every inflammatory mediator. Defining a neuroinflammatory phenotype (NIP) is one way to address this issue, focusing on profiling the changes in inflammatory mediator expression during disease progression. Although many studies have shown that APOE4 induces a detrimental NIP in peripheral inflammation and Aβ-independent neuroinflammation, data for APOE-modulated Aβ-induced neuroinflammation are surprisingly limited. We present data supporting the hypothesis that impaired apoE4 function modulates Aβ-induced effects on inflammatory receptor signaling, including amplification of detrimental (toll-like receptor 4-p38α) and suppression of beneficial (IL-4R-nuclear receptor) pathways. To ultimately develop APOE genotype-specific therapeutics, it is critical that future studies define the dynamic NIP profile and pathways that underlie APOE-modulated chronic neuroinflammation. In this editorial review, we present data supporting the hypothesis that impaired apoE4 function modulates Aβ-induced effects on inflammatory receptor signaling, including amplification of detrimental (TLR4-p38α) and suppression of beneficial (IL-4R-nuclear receptor) pathways, resulting in an adverse NIP that causes neuronal dysfunction. NIP, Neuroinflammatory phenotype; P.I., pro-inflammatory; A.I., anti-inflammatory.
Collapse
Affiliation(s)
- Leon M. Tai
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | - Shivesh Ghura
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | - Kevin P. Koster
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | | | | | - Pinal Kanabar
- UIC Center for Research Informatics University of IllinoisChicagoIllinoisUSA
| | - Nicole Collins
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | - Manel Ben‐Aissa
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| | - Arden Zhengdeng Lei
- UIC Center for Research Informatics University of IllinoisChicagoIllinoisUSA
| | - Neil Bahroos
- UIC Center for Research Informatics University of IllinoisChicagoIllinoisUSA
| | | | - Bill Hendrickson
- UIC Research Resources CenterUniversity of IllinoisChicagoIllinoisUSA
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell BiologyUniversity of IllinoisChicagoIllinoisUSA
| |
Collapse
|
62
|
Abstract
Alzheimer's disease (AD), the most common form of dementia in western societies, is a pathologically and clinically heterogeneous disease with a strong genetic component. The recent advances in high-throughput genome technologies allowing for the rapid analysis of millions of polymorphisms in thousands of subjects has significantly advanced our understanding of the genomic underpinnings of AD susceptibility. During the last 5 years, genome-wide association and whole-exome- and whole-genome sequencing studies have mapped more than 20 disease-associated loci, providing insights into the molecular pathways involved in AD pathogenesis and hinting at potential novel therapeutic targets. This review article summarizes the challenges and opportunities of when using genomic information for the diagnosis and prognosis of AD.
Collapse
Affiliation(s)
- Christiane Reitz
- Sergievsly Center/Taub Institute/Dept. of Neurology, Columbia University, 630 W 168th Street, Rm 19-308, New York, NY 10032, phone: (212) 305-0865, fax: (212) 305-2391
| |
Collapse
|
63
|
Mahmoudi R, Kisserli A, Novella JL, Donvito B, Dramé M, Réveil B, Duret V, Jolly D, Pham BN, Cohen JH. Alzheimer's disease is associated with low density of the long CR1 isoform. Neurobiol Aging 2015; 36:1766.e5-1766.e12. [PMID: 25666996 DOI: 10.1016/j.neurobiolaging.2015.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 10/31/2014] [Accepted: 01/05/2015] [Indexed: 01/21/2023]
Abstract
The long complement receptor type 1 (CR1) isoform, CR1*2 (S), has been identified as being associated with Alzheimer's disease (AD) risk. We aimed to analyze the phenotypic structural and expression aspects (length and density) of CR1 in erythrocytes of 135 Caucasian subjects (100 AD and 35 controls). CR1 length polymorphism was assessed at protein and gene levels using Western blot and high-resolution melting, respectively. CR1 sites on erythrocytes were enumerated by flow cytometry. CR1 gene analysis, spotting the rs6656401 and rs3818361 polymorphisms, was performed by pyrosequencing. The CR1 density was significantly lower in AD patients expressing the CR1*2 isoform compared with the controls (p = 0.001), demonstrating lower expression of CR1 in CR1*2 carriers. Our data suggested the existence of silent CR1 alleles. Finally, rs6656401 and rs3818361 were strongly associated with CR1 length polymorphism (p < 0.0001). These observations indicate that AD susceptibility is associated with the long CR1 isoform (CR1*2), albeit at a lower density, suggesting that AD results from insufficient clearance of plaque deposits rather than increased inflammation.
Collapse
Affiliation(s)
- Rachid Mahmoudi
- Champagne-Ardenne Resource and Research Memory Center (CMRR), Maison Blanche Hospital, Reims University Hospitals, Reims, France; Department of Internal Medicine and Geriatrics, Maison Blanche Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, EA 3797, Reims, France.
| | - Aymric Kisserli
- Department of Immunology, Robert Debré Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, LRN EA 4682, Reims, France
| | - Jean-Luc Novella
- Champagne-Ardenne Resource and Research Memory Center (CMRR), Maison Blanche Hospital, Reims University Hospitals, Reims, France; Department of Internal Medicine and Geriatrics, Maison Blanche Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, EA 3797, Reims, France
| | - Béatrice Donvito
- Department of Immunology, Robert Debré Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, LRN EA 4682, Reims, France
| | - Moustapha Dramé
- Faculty of Medicine, University of Reims Champagne-Ardenne, EA 3797, Reims, France; Department of Research and Innovation, Robert Debré Hospital, Reims University Hospitals, Reims, France
| | - Brigitte Réveil
- Department of Immunology, Robert Debré Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, LRN EA 4682, Reims, France
| | - Valérie Duret
- Department of Immunology, Robert Debré Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, LRN EA 4682, Reims, France
| | - Damien Jolly
- Faculty of Medicine, University of Reims Champagne-Ardenne, EA 3797, Reims, France; Department of Research and Innovation, Robert Debré Hospital, Reims University Hospitals, Reims, France
| | - Bach-Nga Pham
- Department of Immunology, Robert Debré Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, LRN EA 4682, Reims, France
| | - Jacques H Cohen
- Department of Immunology, Robert Debré Hospital, Reims University Hospitals, Reims, France; Faculty of Medicine, University of Reims Champagne-Ardenne, LRN EA 4682, Reims, France
| |
Collapse
|
64
|
Abstract
The small, calcium-sensor protein, calmodulin, is ubiquitously expressed and central to cell function in all cell types. Here the literature linking calmodulin to Alzheimer's disease is reviewed. Several experimentally-verified calmodulin-binding proteins are involved in the formation of amyloid-β plaques including amyloid-β protein precursor, β-secretase, presenilin-1, and ADAM10. Many others possess potential calmodulin-binding domains that remain to be verified. Three calmodulin binding proteins are associated with the formation of neurofibrillary tangles: two kinases (CaMKII, CDK5) and one protein phosphatase (PP2B or calcineurin). Many of the genes recently identified by genome wide association studies and other studies encode proteins that contain putative calmodulin-binding domains but only a couple (e.g., APOE, BIN1) have been experimentally confirmed as calmodulin binding proteins. At least two receptors involved in calcium metabolism and linked to Alzheimer's disease (mAchR; NMDAR) have also been identified as calmodulin-binding proteins. In addition to this, many proteins that are involved in other cellular events intimately associated with Alzheimer's disease including calcium channel function, cholesterol metabolism, neuroinflammation, endocytosis, cell cycle events, and apoptosis have been tentatively or experimentally verified as calmodulin binding proteins. The use of calmodulin as a potential biomarker and as a therapeutic target is discussed.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto at Mississauga, Mississauga, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Kristeen Eshak
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Michael A. Myre
- Center for Human Genetic Research, Richard B. Simches Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
65
|
Hoos MD, Vitek MP, Ridnour LA, Wilson J, Jansen M, Everhart A, Wink DA, Colton CA. The impact of human and mouse differences in NOS2 gene expression on the brain's redox and immune environment. Mol Neurodegener 2014; 9:50. [PMID: 25403885 PMCID: PMC4247207 DOI: 10.1186/1750-1326-9-50] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 10/10/2014] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Mouse models are used in the study of human disease. Despite well-known homologies, the difference in immune response between mice and humans impacts the application of data derived from mice to human disease outcomes. Nitric oxide synthase-2 (NOS2) is a key gene that displays species-specific outcomes via altered regulation of the gene promoter and via post-transcriptional mechanisms in humans that are not found in mice. The resulting levels of NO produced by activation of human NOS2 are different from the levels of NO produced by mouse Nos2. Since both tissue redox environment and immune responsiveness are regulated by the level of NO and its interactions, we investigated the significance of mouse and human differences on brain oxidative stress and on immune activation in HuNOS2tg/mNos2-/- mice that express the entire human NOS2 gene and that lack a functional mNos2 compared to wild type (WT) mice that express normal mNos2. METHODS/RESULTS Similarly to human, brain tissue from HuNOS2tg/mNos2-/- mice showed the presence of a NOS2 gene 3'UTR binding site. We also identified miRNA-939, the binding partner for this site, in mouse brain lysates and further demonstrated reduced levels of nitric oxide (NO) typical of the human immune response on injection with lipopolysaccharide (LPS). HuNOS2tg/mNos2-/- brain samples were probed for characteristic differences in redox and immune gene profiles compared to WT mice using gene arrays. Selected genes were also compared against mNos2-/- brain lysates. Reconstitution of the human NOS2 gene significantly altered genes that encode multiple anti-oxidant proteins, oxidases, DNA repair, mitochondrial proteins and redox regulated immune proteins. Expression levels of typical pro-inflammatory, anti-inflammatory and chemokine genes were not significantly different with the exception of increased TNFα and Ccr1 mRNA expression in the HuNOS2tg/mNos2-/- mice compared to WT or mNos2-/- mice. CONCLUSIONS NO is a principle factor in establishing the tissue redox environment and changes in NO levels impact oxidative stress and immunity, both of which are primary characteristics of neurodegenerative diseases. The HuNOS2tg/mNos2-/- mice provide a potentially useful mechanism to address critical species- specific immune differences that can impact the study of human diseases.
Collapse
Affiliation(s)
- Michael D Hoos
- />Department of Neurosurgery, Stonybrook Health Sciences, Stony Brook, NY 11794 USA
| | - Michael P Vitek
- />Department of Neurology, Duke University Medical Center, Durham, NC 27710 USA
| | - Lisa A Ridnour
- />Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Joan Wilson
- />Department of Neurology, Duke University Medical Center, Durham, NC 27710 USA
| | - Marilyn Jansen
- />Department of Neurology, Duke University Medical Center, Durham, NC 27710 USA
| | - Angela Everhart
- />Department of Neurology, Duke University Medical Center, Durham, NC 27710 USA
| | - David A Wink
- />Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Carol A Colton
- />Department of Neurology, Duke University Medical Center, Durham, NC 27710 USA
| |
Collapse
|
66
|
Paterson RW, Toombs J, Slattery CF, Schott JM, Zetterberg H. Biomarker modelling of early molecular changes in Alzheimer's disease. Mol Diagn Ther 2014; 18:213-27. [PMID: 24281842 DOI: 10.1007/s40291-013-0069-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The preclinical phase of Alzheimer's disease (AD) occurs years, possibly decades, before the onset of clinical symptoms. Being able to detect the very earliest stages of AD is critical to improving understanding of AD biology, and identifying individuals at greatest risk of developing clinical symptoms with a view to treating AD pathophysiology before irreversible neurodegeneration occurs. Studies of dominantly inherited AD families and longitudinal studies of sporadic AD have contributed to knowledge of the earliest AD biomarkers. Here we appraise this evidence before reviewing novel, particularly fluid, biomarkers that may provide insights into AD pathogenesis and relate these to existing hypothetical disease models.
Collapse
Affiliation(s)
- Ross W Paterson
- Dementia Research Centre, Department of Neurodegeneration, UCL Institute of Neurology, London, UK,
| | | | | | | | | |
Collapse
|
67
|
Stilling RM, Benito E, Gertig M, Barth J, Capece V, Burkhardt S, Bonn S, Fischer A. De-regulation of gene expression and alternative splicing affects distinct cellular pathways in the aging hippocampus. Front Cell Neurosci 2014; 8:373. [PMID: 25431548 PMCID: PMC4230043 DOI: 10.3389/fncel.2014.00373] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 10/21/2014] [Indexed: 01/20/2023] Open
Abstract
Aging is accompanied by gradually increasing impairment of cognitive abilities and constitutes the main risk factor of neurodegenerative conditions like Alzheimer's disease (AD). The underlying mechanisms are however not well understood. Here we analyze the hippocampal transcriptome of young adult mice and two groups of mice at advanced age using RNA sequencing. This approach enabled us to test differential expression of coding and non-coding transcripts, as well as differential splicing and RNA editing. We report a specific age-associated gene expression signature that is associated with major genetic risk factors for late-onset AD (LOAD). This signature is dominated by neuroinflammatory processes, specifically activation of the complement system at the level of increased gene expression, while de-regulation of neuronal plasticity appears to be mediated by compromised RNA splicing.
Collapse
Affiliation(s)
- Roman M Stilling
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen Göttingen, Germany ; Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Eva Benito
- Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Michael Gertig
- Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Jonas Barth
- Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Vincenzo Capece
- Research Group for Computational Analysis of Biological Networks, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Susanne Burkhardt
- Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Stefan Bonn
- Research Group for Computational Analysis of Biological Networks, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| | - Andre Fischer
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen Göttingen, Germany ; Research Group for Epigenetics in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Göttingen Göttingen, Germany
| |
Collapse
|
68
|
Ahmed S, Kemp MW, Payne MS, Kallapur SG, Stock SJ, Marsh HC, Jobe AH, Newnham JP, Spiller OB. Comparison of complement activity in adult and preterm sheep serum. Am J Reprod Immunol 2014; 73:232-41. [PMID: 25046333 DOI: 10.1111/aji.12299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 07/02/2014] [Indexed: 01/25/2023] Open
Abstract
PROBLEM Functional complement activity is routinely measured utilizing rabbit antibody-sensitized sheep erythrocytes. Due to complement inhibitor expression on erythrocytes, the development of an alternative method to measure complement function in sheep serum was required. METHOD OF STUDY Several species of target erythrocyte and sensitizing antibody were investigated for improved measurement of complement function testing. RESULTS AND CONCLUSION Guinea pig erythrocytes were identified as the optimal target, although sensitizing them with rabbit antiguinea pig erythrocyte antibody did not enhance the lysis by maternal sheep serum. In contrast, preterm neonatal sheep serum was unable to efficiently lyse guinea pig erythrocytes unless pre-sensitized with antibody. Further investigation revealed that maternal serum contained high levels of antibodies that cross-reacted with guinea pig and rabbit erythrocytes, while no cross-reacting antierythrocyte antibodies were found in preterm neonatal serum. Therefore, unlike primates, rabbits, and guinea pigs, no transplacental transfer of maternal IgG to foetal sheep occurs. Use of exogenous complement regulators is often used to dissect the contribution of complement to disease pathogenesis; however, we found that while full-length soluble human complement receptor 1 (sCR1, CDX-1135) was able to inhibit lysis of guinea pig erythrocytes by human and rat serum, no inhibition of sheep serum could be observed. Investigation of complement contribution to disease pathogenesis in the future will require the identification of an inhibitor that is effective against sheep complement.
Collapse
Affiliation(s)
- Shatha Ahmed
- School of Medicine, Institute of Molecular and Experimental Medicine, Cardiff University, University Hospital of Wales, Cardiff, UK; Department of Pathology, Nineveh College of Medicine, University of Mosul, Mosul, Iraq
| | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Luo J, Li S, Qin X, Song L, Peng Q, Chen S, Xie Y, Xie L, Li T, He Y, Deng Y, Wang J, Zeng Z. Meta-analysis of the association between CR1 polymorphisms and risk of late-onset Alzheimer's disease. Neurosci Lett 2014; 578:165-70. [PMID: 24996192 DOI: 10.1016/j.neulet.2014.06.055] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 06/14/2014] [Accepted: 06/24/2014] [Indexed: 10/25/2022]
Abstract
CR1 polymorphisms have been reported to be associated with late-onset Alzheimer's disease (LOAD) susceptibility. The findings of these studies, however, have been inconsistent. Therefore, we performed a meta-analysis to assess the association between CR1 variants and LOAD susceptibility. We retrieved all relevant studies of the associations between CR1 polymorphisms and the susceptibility to LOAD for the period up to March 30, 2014. The strength of the association between CR1 polymorphisms and LOAD risk was estimated by odds ratios (ORs) and their 95% confidence intervals (CIs). A total of 6 articles were eventually identified with 2752 LOAD cases and 2313 controls for the rs6656401 polymorphism, and 4 studies containing 2547 LOAD cases and 2338 controls were included for the rs3818361 polymorphism. Overall, the pooled data showed that the CR1 rs6656401 polymorphism was significantly associated with LOAD risk in the overall population (A vs. G: OR=1.32, 95%CI=1.17-1.50, P=0.000; AG+AA vs. GG: OR=1.39, 95%CI=1.20-1.61, P=0.000). With respect to the CR1 rs3818361 polymorphism, a statistically significant increased LOAD risk was observed in the overall population (T vs. C: OR=1.24, 95% CI=1.13-1.37, P=0.000; TT+TC vs. CC: OR=1.30, 95% CI=1.15-1.46, P=0.000; TT vs. TC+CC: OR=1.35, 95% CI=1.06-1.71, P=0.014). This meta-analysis demonstrated significant associations of both the CR1 rs6656401 and CR1 rs3818361 polymorphisms with LOAD susceptibility.
Collapse
Affiliation(s)
- Jingrong Luo
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Shan Li
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Xue Qin
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Liuying Song
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Qiliu Peng
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Siyuan Chen
- Guangxi University of Chinese Medicine, Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Yantong Xie
- Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Li Xie
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Taijie Li
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Yu He
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Yan Deng
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Jian Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Zhiyu Zeng
- Vasculocardiology Department, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning 530021, China.
| |
Collapse
|
70
|
Kanekiyo T, Bu G. The low-density lipoprotein receptor-related protein 1 and amyloid-β clearance in Alzheimer's disease. Front Aging Neurosci 2014; 6:93. [PMID: 24904407 PMCID: PMC4033011 DOI: 10.3389/fnagi.2014.00093] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 04/28/2014] [Indexed: 11/24/2022] Open
Abstract
Accumulation and aggregation of amyloid-β (Aβ) peptides in the brain trigger the development of progressive neurodegeneration and dementia associated with Alzheimer’s disease (AD). Perturbation in Aβ clearance, rather than Aβ production, is likely the cause of sporadic, late-onset AD, which accounts for the majority of AD cases. Since cellular uptake and subsequent degradation constitute a major Aβ clearance pathway, the receptor-mediated endocytosis of Aβ has been intensely investigated. Among Aβ receptors, the low-density lipoprotein receptor-related protein 1 (LRP1) is one of the most studied receptors. LRP1 is a large endocytic receptor for more than 40 ligands, including apolipoprotein E, α2-macroglobulin and Aβ. Emerging in vitro and in vivo evidence demonstrates that LRP1 is critically involved in brain Aβ clearance. LRP1 is highly expressed in a variety of cell types in the brain including neurons, vascular cells and glial cells, where LRP1 functions to maintain brain homeostasis and control Aβ metabolism. LRP1-mediated endocytosis regulates cellular Aβ uptake by binding to Aβ either directly or indirectly through its co-receptors or ligands. Furthermore, LRP1 regulates several signaling pathways, which also likely influences Aβ endocytic pathways. In this review, we discuss how LRP1 regulates the brain Aβ clearance and how this unique endocytic receptor participates in AD pathogenesis. Understanding of the mechanisms underlying LRP1-mediated Aβ clearance should enable the rational design of novel diagnostic and therapeutic strategies for AD.
Collapse
Affiliation(s)
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville FL, USA
| |
Collapse
|
71
|
Shinohara M, Sato N, Shimamura M, Kurinami H, Hamasaki T, Chatterjee A, Rakugi H, Morishita R. Possible modification of Alzheimer's disease by statins in midlife: interactions with genetic and non-genetic risk factors. Front Aging Neurosci 2014; 6:71. [PMID: 24795626 PMCID: PMC4005936 DOI: 10.3389/fnagi.2014.00071] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 03/30/2014] [Indexed: 12/28/2022] Open
Abstract
The benefits of statins, commonly prescribed for hypercholesterolemia, in treating Alzheimer's disease (AD) have not yet been fully established. A recent randomized clinical trial did not show any therapeutic effects of two statins on cognitive function in AD. Interestingly, however, the results of the Rotterdam study, one of the largest prospective cohort studies, showed reduced risk of AD in statin users. Based on the current understanding of statin actions and AD pathogenesis, it is still worth exploring whether statins can prevent AD when administered decades before the onset of AD or from midlife. This review discusses the possible beneficial effects of statins, drawn from previous clinical observations, pathogenic mechanisms, which include β-amyloid (Aβ) and tau metabolism, genetic and non-genetic risk factors (apolipoprotein E, cholesterol, sex, hypertension, and diabetes), and other clinical features (vascular dysfunction and oxidative and inflammatory stress) of AD. These findings suggest that administration of statins in midlife might prevent AD in late life by modifying genetic and non-genetic risk factors for AD. It should be clarified whether statins inhibit Aβ accumulation, tau pathological features, and brain atrophy in humans. To answer this question, a randomized controlled study using amyloid positron emission tomography (PET), tau-PET, and magnetic resonance imaging would be useful. This clinical evaluation could help us to overcome this devastating disease.
Collapse
Affiliation(s)
- Mitsuru Shinohara
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Naoyuki Sato
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Munehisa Shimamura
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University Office for University-Industry CollaborationSuita, Japan
| | - Hitomi Kurinami
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University Office for University-Industry CollaborationSuita, Japan
| | - Toshimitsu Hamasaki
- Department of Biomedical Statistics, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Amarnath Chatterjee
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Hiromi Rakugi
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
| |
Collapse
|
72
|
Abstract
Alzheimer’s disease (AD) is a complex and heterogeneous neurodegenerative disorder, classified as either early onset (under 65 years of age), or late onset (over 65 years of age). Three main genes are involved in early onset AD: amyloid precursor protein (APP), presenilin 1 (PSEN1), and presenilin 2 (PSEN2). The apolipoprotein E (APOE) E4 allele has been found to be a main risk factor for late-onset Alzheimer’s disease. Additionally, genome-wide association studies (GWASs) have identified several genes that might be potential risk factors for AD, including clusterin (CLU), complement receptor 1 (CR1), phosphatidylinositol binding clathrin assembly protein (PICALM), and sortilin-related receptor (SORL1). Recent studies have discovered additional novel genes that might be involved in late-onset AD, such as triggering receptor expressed on myeloid cells 2 (TREM2) and cluster of differentiation 33 (CD33). Identification of new AD-related genes is important for better understanding of the pathomechanisms leading to neurodegeneration. Since the differential diagnoses of neurodegenerative disorders are difficult, especially in the early stages, genetic testing is essential for diagnostic processes. Next-generation sequencing studies have been successfully used for detecting mutations, monitoring the epigenetic changes, and analyzing transcriptomes. These studies may be a promising approach toward understanding the complete genetic mechanisms of diverse genetic disorders such as AD.
Collapse
Affiliation(s)
- Eva Bagyinszky
- Department of BioNano Technology Gachon University, Gyeonggi-do, South Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Seong Soo A An
- Department of BioNano Technology Gachon University, Gyeonggi-do, South Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University Budang Hospital, Gyeonggi-do, South Korea
| |
Collapse
|
73
|
Yasuda M, Tanaka Y, Ryu M, Tsuda S, Nakazawa T. RNA sequence reveals mouse retinal transcriptome changes early after axonal injury. PLoS One 2014; 9:e93258. [PMID: 24676137 PMCID: PMC3968129 DOI: 10.1371/journal.pone.0093258] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 03/04/2014] [Indexed: 12/23/2022] Open
Abstract
Glaucoma is an ocular disease characterized by progressive retinal ganglion cell (RGC) death caused by axonal injury. However, the underlying mechanisms involved in RGC death remain unclear. In this study, we investigated changes in the transcriptome profile following axonal injury in mice (C57BL/6) with RNA sequencing (RNA-seq) technology. The experiment group underwent an optic nerve crush (ONC) procedure to induce axonal injury in the right eye, and the control group underwent a sham procedure. Two days later, we extracted the retinas and performed RNA-seq and a pathway analysis. We identified 177 differentially expressed genes with RNA-seq, notably the endoplasmic reticulum (ER) stress-related genes Atf3, Atf4, Atf5, Chac1, Chop, Egr1 and Trb3, which were significantly upregulated. The pathway analysis revealed that ATF4 was the most significant upstream regulator. The antioxidative response-related genes Hmox1 and Srxn1, as well as the immune response-related genes C1qa, C1qb and C1qc, were also significantly upregulated. To our knowledge, this is the first reported RNA-seq investigation of the retinal transcriptome and molecular pathways in the early stages after axonal injury. Our results indicated that ER stress plays a key role under these conditions. Furthermore, the antioxidative defense and immune responses occurred concurrently in the early stages after axonal injury. We believe that our study will lead to a better understanding of and insight into the molecular mechanisms underlying RGC death after axonal injury.
Collapse
Affiliation(s)
- Masayuki Yasuda
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuji Tanaka
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Morin Ryu
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoru Tsuda
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- * E-mail:
| |
Collapse
|
74
|
Abstract
Over the past decade, research aiming to disentangle the genetic underpinnings of late-onset Alzheimer’s disease has mostly focused on the identification of common variants through genome-wide association studies. The identification of several new susceptibility genes through these efforts has reinforced the importance of amyloid precursor protein and tau metabolism in the cause of the disease and has implicated immune response, inflammation, lipid metabolism, endocytosis/intracellular trafficking, and cell migration in the cause of the disease. Ongoing and future large-scale genome-wide association studies, translational studies, and next-generation whole genome or whole exome sequencing efforts, hold the promise to map the specific causative variants in these genes, to identify several additional risk variants, including rare and structural variants, and to identify novel targets for genetic testing, prevention, and treatment.
Collapse
Affiliation(s)
- Giuseppe Tosto
- />Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY USA
| | - Christiane Reitz
- />Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY USA
- />Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY USA
- />Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032 USA
| |
Collapse
|
75
|
Doens D, Fernández PL. Microglia receptors and their implications in the response to amyloid β for Alzheimer's disease pathogenesis. J Neuroinflammation 2014; 11:48. [PMID: 24625061 PMCID: PMC3975152 DOI: 10.1186/1742-2094-11-48] [Citation(s) in RCA: 237] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 02/24/2014] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a major public health problem with substantial economic and social impacts around the world. The hallmarks of AD pathogenesis include deposition of amyloid β (Aβ), neurofibrillary tangles, and neuroinflammation. For many years, research has been focused on Aβ accumulation in senile plaques, as these aggregations were perceived as the main cause of the neurodegeneration found in AD. However, increasing evidence suggests that inflammation also plays a critical role in the pathogenesis of AD. Microglia cells are the resident macrophages of the brain and act as the first line of defense in the central nervous system. In AD, microglia play a dual role in disease progression, being essential for clearing Aβ deposits and releasing cytotoxic mediators. Aβ activates microglia through a variety of innate immune receptors expressed on these cells. The mechanisms through which amyloid deposits provoke an inflammatory response are not fully understood, but it is believed that these receptors cooperate in the recognition, internalization, and clearance of Aβ and in cell activation. In this review, we discuss the role of several receptors expressed on microglia in Aβ recognition, uptake, and signaling, and their implications for AD pathogenesis.
Collapse
Affiliation(s)
- Deborah Doens
- Centro de Biología Molecular y Celular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), Edificio 219, Clayton, Ciudad del Saber, República de Panamá
- Department of Biotechnology, Acharya Nagarjuna University, Guntur, India
| | - Patricia L Fernández
- Centro de Biología Molecular y Celular de Enfermedades, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), Edificio 219, Clayton, Ciudad del Saber, República de Panamá
| |
Collapse
|
76
|
Reitz C, Mayeux R. Alzheimer disease: epidemiology, diagnostic criteria, risk factors and biomarkers. Biochem Pharmacol 2014; 88:640-51. [PMID: 24398425 DOI: 10.1016/j.bcp.2013.12.024] [Citation(s) in RCA: 766] [Impact Index Per Article: 76.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 12/20/2013] [Accepted: 12/20/2013] [Indexed: 12/20/2022]
Abstract
The global prevalence of dementia is as high as 24 million, and has been predicted to quadruple by the year 2050. In the US alone, Alzheimer disease (AD) - the most frequent cause of dementia characterized by a progressive decline in cognitive function in particular the memory domain - causes estimated health-care costs of $ 172 billion per year. Key neuropathological hallmarks of the AD brain are diffuse and neuritic extracellular amyloid plaques - often surrounded by dystrophic neurites - and intracellular neurofibrillary tangles. These pathological changes are frequently accompanied by reactive microgliosis and loss of neurons, white matter and synapses. The etiological mechanisms underlying these neuropathological changes remain unclear, but are probably caused by both environmental and genetic factors. In this review article, we provide an overview of the epidemiology of AD, review the biomarkers that may be used for risk assessment and in diagnosis, and give suggestions for future research.
Collapse
Affiliation(s)
- Christiane Reitz
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, United States; Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, United States; Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, United States; Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, United States; Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, United States; Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, NY, United States; Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, United States.
| |
Collapse
|
77
|
McGeer PL, McGeer EG. The amyloid cascade-inflammatory hypothesis of Alzheimer disease: implications for therapy. Acta Neuropathol 2013; 126:479-97. [PMID: 24052108 DOI: 10.1007/s00401-013-1177-7] [Citation(s) in RCA: 302] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 09/03/2013] [Accepted: 09/05/2013] [Indexed: 12/14/2022]
Abstract
The amyloid cascade hypothesis is widely accepted as the centerpiece of Alzheimer disease (AD) pathogenesis. It proposes that abnormal production of beta amyloid protein (Abeta) is the cause of AD and that the neurotoxicity is due to Abeta itself or its oligomeric forms. We suggest that this, in itself, cannot be the cause of AD because demonstrating such toxicity requires micromolar concentrations of these Abeta forms, while their levels in brain are a million times lower in the picomolar range. AD probably results from the inflammatory response induced by extracellular Abeta deposits, which later become enhanced by aggregates of tau. The inflammatory response, which is driven by activated microglia, increases over time as the disease progresses. Disease-modifying therapeutic attempts to date have failed and may continue to do so as long as the central role of inflammation is not taken into account. Multiple epidemiological and animal model studies show that NSAIDs, the most widely used antiinflammatory agents, have a substantial sparing effect on AD. These studies provide a proof of concept regarding the anti-inflammatory approach to disease modification. Biomarker studies have indicated that early intervention may be necessary. They have established that disease onset occurs more than a decade before it becomes clinically evident. By combining biomarker and pathological data, it is possible to define six phases of disease development, each separated by about 5 years. Phase one can be identified by decreases in Abeta in the CSF, phase 2 by increases of tau in the CSF plus clear evidence of Abeta brain deposits by PET scanning, phase 3 by slight decreases in brain metabolic rate by PET-FDG scanning, phase 4 by slight decreases in brain volume by MRI scanning plus minimal cognitive impairment, phase 5 by increased scanning abnormalities plus clinical diagnosis of AD, and phase 6 by advanced AD requiring institutional care. Utilization of antiinflammatory agents early in the disease process remains an overlooked therapeutic opportunity. Such agents, while not preventative, have the advantage of being able to inhibit the consequences of both Abeta and tau aggregation. Since there is more than a decade between disease onset and cognitive decline, a window of opportunity exists to introduce truly effective disease-modifying regimens. Taking advantage of this opportunity is the challenge for the future.
Collapse
Affiliation(s)
- Patrick L McGeer
- Kinsmen Laboratory of Neurological Research, University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T1Z3, Canada,
| | | |
Collapse
|
78
|
Melhorn MI, Brodsky AS, Estanislau J, Khoory JA, Illigens B, Hamachi I, Kurishita Y, Fraser AD, Nicholson-Weller A, Dolmatova E, Duffy HS, Ghiran IC. CR1-mediated ATP release by human red blood cells promotes CR1 clustering and modulates the immune transfer process. J Biol Chem 2013; 288:31139-53. [PMID: 24022490 DOI: 10.1074/jbc.m113.486035] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Humans and other higher primates are unique among mammals in using complement receptor 1 (CR1, CD35) on red blood cells (RBC) to ligate complement-tagged inflammatory particles (immune complexes, apoptotic/necrotic debris, and microbes) in the circulation for quiet transport to the sinusoids of spleen and liver where resident macrophages remove the particles, but allow the RBC to return unharmed to the circulation. This process is called immune-adherence clearance. In this study we found using luminometric- and fluorescence-based methods that ligation of CR1 on human RBC promotes ATP release. Our data show that CR1-mediated ATP release does not depend on Ca(2+) or enzymes previously shown to mediate an increase in membrane deformability promoted by CR1 ligation. Furthermore, ATP release following CR1 ligation increases the mobility of the lipid fraction of RBC membranes, which in turn facilitates CR1 clustering, and thereby enhances the binding avidity of complement-opsonized particles to the RBC CR1. Finally, we have found that RBC-derived ATP has a stimulatory effect on phagocytosis of immune-adherent immune complexes.
Collapse
|
79
|
Blockage of CR1 prevents activation of rodent microglia. Neurobiol Dis 2013; 54:139-49. [PMID: 23454195 DOI: 10.1016/j.nbd.2013.02.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 01/31/2013] [Accepted: 02/19/2013] [Indexed: 12/22/2022] Open
Abstract
The importance of the complement system in Alzheimer's disease (AD) pathogenesis has been emphasized through recent genome wide association studies. However, the cellular and molecular role of these complement proteins is not fully understood. Microglia express complement receptors and the activation of specific receptors may increase Aβ clearance and reduce neurodegeneration. Here, we investigated the contribution of complement receptor 1 (CR1), the second most significant hit in GWAS studies, on microglia to neuronal damage. We show that microglia displaying an activated phenotype demonstrate an increase in CR1 expression. We also provide evidence that activation of microglial CR1 was detrimental to neurons and this correlated with an increase in microglial intracellular superoxide generation, and tumour necrosis factor-α (TNFα) and interleukin-1 β (IL-1β) secretion. Amyloid-β 42 (Aβ1-42)-treated microglia displayed an increased ability to phagocytose dextran beads following antibody blockage of CR1 but a decreased capacity to phagocytose fluorescent-tagged Aβ1-42. Together, these results indicate that microglial CR1 plays a role in the neuronal death observed in AD and investigating this further may provide a possible strategy to control neurotoxicity in the AD brain.
Collapse
|
80
|
Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, Cruchaga C, Sassi C, Kauwe JSK, Younkin S, Hazrati L, Collinge J, Pocock J, Lashley T, Williams J, Lambert JC, Amouyel P, Goate A, Rademakers R, Morgan K, Powell J, St George-Hyslop P, Singleton A, Hardy J. TREM2 variants in Alzheimer's disease. N Engl J Med 2013; 368:117-27. [PMID: 23150934 PMCID: PMC3631573 DOI: 10.1056/nejmoa1211851] [Citation(s) in RCA: 2154] [Impact Index Per Article: 195.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Homozygous loss-of-function mutations in TREM2, encoding the triggering receptor expressed on myeloid cells 2 protein, have previously been associated with an autosomal recessive form of early-onset dementia. METHODS We used genome, exome, and Sanger sequencing to analyze the genetic variability in TREM2 in a series of 1092 patients with Alzheimer's disease and 1107 controls (the discovery set). We then performed a meta-analysis on imputed data for the TREM2 variant rs75932628 (predicted to cause a R47H substitution) from three genomewide association studies of Alzheimer's disease and tested for the association of the variant with disease. We genotyped the R47H variant in an additional 1887 cases and 4061 controls. We then assayed the expression of TREM2 across different regions of the human brain and identified genes that are differentially expressed in a mouse model of Alzheimer's disease and in control mice. RESULTS We found significantly more variants in exon 2 of TREM2 in patients with Alzheimer's disease than in controls in the discovery set (P=0.02). There were 22 variant alleles in 1092 patients with Alzheimer's disease and 5 variant alleles in 1107 controls (P<0.001). The most commonly associated variant, rs75932628 (encoding R47H), showed highly significant association with Alzheimer's disease (P<0.001). Meta-analysis of rs75932628 genotypes imputed from genomewide association studies confirmed this association (P=0.002), as did direct genotyping of an additional series of 1887 patients with Alzheimer's disease and 4061 controls (P<0.001). Trem2 expression differed between control mice and a mouse model of Alzheimer's disease. CONCLUSIONS Heterozygous rare variants in TREM2 are associated with a significant increase in the risk of Alzheimer's disease. (Funded by Alzheimer's Research UK and others.).
Collapse
Affiliation(s)
- Rita Guerreiro
- University College London (UCL) Institute of Neurology, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Killick R, Hughes TR, Morgan BP, Lovestone S. Deletion of Crry, the murine ortholog of the sporadic Alzheimer's disease risk gene CR1, impacts tau phosphorylation and brain CFH. Neurosci Lett 2012; 533:96-9. [PMID: 23153828 PMCID: PMC3556777 DOI: 10.1016/j.neulet.2012.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 10/25/2012] [Accepted: 11/05/2012] [Indexed: 01/09/2023]
Abstract
Large-scale genome-wide SNP association studies have identified an association between variants of CR1, the gene encoding complement component receptor 1, and the sporadic form of Alzheimer's disease. The role of CR1 and the complement system in Alzheimer's disease remains far from clear. In rodents the closest ortholog of CR1 is the Crry gene (Cr1-related protein Y). To begin to explore its role in Alzheimer's disease we examined hippocampal lysates from Crry−/− mice and age matched controls by immunoblotting. We measured complement factor H, a component of the complement system and biomarker for Alzheimer's disease progression, and tau phosphorylation at the serine 235 site, hyperphosphorylated forms of tau being a defining neuropathological hallmark of the disease. We found that levels of CFH and of tau phosphorylation at serine 235 were strongly and significantly reduced in Crry−/− samples. These observations provide a starting point for further attempts to determine the role of CR1 in the neuropathological process driving Alzheimer's disease.
Collapse
Affiliation(s)
- R Killick
- King's College London, Institute of Psychiatry, De Crespigny Park, Denmark Hill, London SE5 8AF, UK.
| | | | | | | |
Collapse
|
82
|
Schellenberg GD, Montine TJ. The genetics and neuropathology of Alzheimer's disease. Acta Neuropathol 2012; 124:305-23. [PMID: 22618995 DOI: 10.1007/s00401-012-0996-2] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 05/07/2012] [Accepted: 05/08/2012] [Indexed: 02/07/2023]
Abstract
Here we review the genetic causes and risks for Alzheimer's disease (AD). Early work identified mutations in three genes that cause AD: APP, PSEN1 and PSEN2. Although mutations in these genes are rare causes of AD, their discovery had a major impact on our understanding of molecular mechanisms of AD. Early work also revealed the ε4 allele of the APOE as a strong risk factor for AD. Subsequently, SORL1 also was identified as an AD risk gene. More recently, advances in our knowledge of the human genome, made possible by technological advances and methods to analyze genomic data, permit systematic identification of genes that contribute to AD risk. This work, so far accomplished through single nucleotide polymorphism arrays, has revealed nine new genes implicated in AD risk (ABCA7, BIN1, CD33, CD2AP, CLU, CR1, EPHA1, MS4A4E/MS4A6A, and PICALM). We review the relationship between these mutations and genetic variants and the neuropathologic features of AD and related disorders. Together, these discoveries point toward a new era in neurodegenerative disease research that impacts not only AD but also related illnesses that produce cognitive and behavioral deficits.
Collapse
Affiliation(s)
- Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6100, USA.
| | | |
Collapse
|
83
|
Gou SJ, Yuan J, Chen M, Yu F, Zhao MH. Circulating complement activation in patients with anti-neutrophil cytoplasmic antibody-associated vasculitis. Kidney Int 2012; 83:129-37. [PMID: 22913983 DOI: 10.1038/ki.2012.313] [Citation(s) in RCA: 190] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Studies in animal models suggest that complement activation is crucial in the pathogenesis of anti-neutrophil cytoplasmic antibody-associated vasculitis (AAV). Here we investigate the circulating complement activation profile of 66 patients with active stage AAV compared to that of 54 patients with AAV in remission. Plasma levels of C3a, C5a, soluble C5b-9, and Bb, all determined by enzyme-linked immunosorbent assay, were significantly higher in active stage than in remission of AAV, while plasma levels of properdin were significantly lower in the former than the latter disease stage. There was no significant difference in the plasma levels of C4d between active stage and remission. The plasma level of Bb in patients with active AAV significantly correlated with the proportion of total and cellular crescents in the renal biopsy, the erythrocyte sedimentation rate, and the Birmingham Vasculitis Activity Scores. Thus, systemic activation of complement by the alternative pathway takes place in human AAV. Circulating Bb might be a useful biomarker in assessing disease activity of AAV.
Collapse
Affiliation(s)
- Shen-Ju Gou
- Renal Division, Department of Medicine, Peking University First Hospital, Institute of Nephrology, Peking University, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, China
| | | | | | | | | |
Collapse
|
84
|
Microglia, Alzheimer's disease, and complement. Int J Alzheimers Dis 2012; 2012:983640. [PMID: 22957298 PMCID: PMC3432348 DOI: 10.1155/2012/983640] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 05/01/2012] [Accepted: 05/07/2012] [Indexed: 02/02/2023] Open
Abstract
Microglia, the immune cell of the brain, are implicated in cascades leading to neuronal loss and cognitive decline in Alzheimer's disease (AD). Recent genome-wide association studies have indicated a number of risk factors for the development of late-onset AD. Two of these risk factors are an altered immune response and polymorphisms in complement receptor 1. In view of these findings, we discuss how complement signalling in the AD brain and microglial responses in AD intersect. Dysregulation of the complement cascade, either by changes in receptor expression, enhanced activation of different complement pathways or imbalances between complement factor production and complement cascade inhibitors may all contribute to the involvement of complement in AD. Altered complement signalling may reduce the ability of microglia to phagocytose apoptotic cells and clear amyloid beta peptides, modulate the expression by microglia of complement components and receptors, promote complement factor production by plaque-associated cytokines derived from activated microglia and astrocytes, and disrupt complement inhibitor production. The evidence presented here indicates that microglia in AD are influenced by complement factors to adopt protective or harmful phenotypes and the challenge ahead lies in understanding how this can be manipulated to therapeutic advantage to treat late onset AD.
Collapse
|
85
|
Linnartz B, Neumann H. Microglial activatory (immunoreceptor tyrosine-based activation motif)- and inhibitory (immunoreceptor tyrosine-based inhibition motif)-signaling receptors for recognition of the neuronal glycocalyx. Glia 2012; 61:37-46. [PMID: 22615186 DOI: 10.1002/glia.22359] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 04/30/2012] [Indexed: 11/09/2022]
Abstract
Microglia sense intact or lesioned cells of the central nervous system (CNS) and respond accordingly. To fulfill this task, microglia express a whole set of recognition receptors. Fc receptors and DAP12 (TYROBP)-associated receptors such as microglial triggering receptor expressed on myeloid cells-2 (TREM2) and the complement receptor-3 (CR3, CD11b/CD18) trigger the immunoreceptor tyrosine-based activation motif (ITAM)-signaling cascade, resulting in microglial activation, migration, and phagocytosis. Those receptors are counter-regulated by immunoreceptor tyrosine-based inhibition motif (ITIM)-signaling receptors, such as sialic acid-binding immunoglobulin superfamily lectins (Siglecs). Siglecs recognize the sialic acid cap of healthy neurons thus leading to an ITIM signaling that turns down microglial immune responses and phagocytosis. In contrast, desialylated neuronal processes are phagocytosed by microglial CR3 signaling via an adaptor protein containing an ITAM. Thus, the aberrant terminal glycosylation of neuronal surface glycoproteins and glycolipids could serve as a flag for microglia, which display a multitude of diverse carbohydrate-binding receptors that monitor the neuronal physical condition and respond via their ITIM- or ITAM-signaling cascade accordingly.
Collapse
Affiliation(s)
- Bettina Linnartz
- Neural Regeneration, Institute of Reconstructive Neurobiology, University Hospital Bonn, University Bonn, 53127 Bonn, Germany
| | | |
Collapse
|
86
|
Complement activation as a biomarker for Alzheimer's disease. Immunobiology 2011; 217:204-15. [PMID: 21856034 DOI: 10.1016/j.imbio.2011.07.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2011] [Revised: 06/22/2011] [Accepted: 07/18/2011] [Indexed: 01/31/2023]
Abstract
There is increasing evidence from genetic, immunohistochemical, proteomic and epidemiological studies as well as in model systems that complement activation has an important role in the pathogenesis of Alzheimer's disease (AD). The complement cascade is an essential element of the innate immune response. In the brain complement proteins are integral components of amyloid plaques and complement activation occurs at the earliest stage of the disease. The complement cascade has been implicated as a protective mechanism in the clearance of amyloid, and in a causal role through chronic activation of the inflammatory response. In this review we discuss the potential for complement activation to act as a biomarker for AD at several stages in the disease process. An accurate biomarker that has sufficient predictive, diagnostic and prognostic value would provide a significant opportunity to develop and test for effective novel therapies in the treatment of AD.
Collapse
|