Genetic variability in response to Aβ deposition influences Alzheimer’s risk

Genetic analysis of late-onset Alzheimer’s disease risk has previously identified a network of largely microglial genes that form a transcriptional network. In transgenic mouse models of amyloid deposition we have previously shown that the expression of many of the mouse orthologs of these genes are co-ordinately up-regulated by amyloid deposition. Here we investigate whether systematic analysis of other members of this mouse amyloid-responsive network predicts other Alzheimer’s risk loci. This statistical comparison of the mouse amyloid-response network with Alzheimer’s disease genome-wide association studies identifies 5 other genetic risk loci for the disease (OAS1, CXCL10, LAPTM5, ITGAM and LILRB4). This work suggests that genetic variability in the microglial response to amyloid deposition is a major determinant for Alzheimer’s risk. One Sentence Summary Identification of 5 new risk loci for Alzheimer’s by statistical comparison of mouse Aβ microglial response with gene-based SNPs from human GWAS

[1]  Young-Joon Kim,et al.  OAS1 and OAS3 negatively regulate the expression of chemokines and interferon-responsive genes in human macrophages , 2019, BMB reports.

[2]  Hans-Ulrich Klein,et al.  Descriptor : A multi-omic atlas of the human frontal cortex for aging and Alzheimer ’ s disease research , 2018 .

[3]  R. Ransohoff,et al.  The Trem2 R47H variant confers loss-of-function-like phenotypes in Alzheimer’s disease , 2018, Molecular Neurodegeneration.

[4]  J. Gallacher,et al.  Meta-analysis of genetic association with diagnosed Alzheimer’s disease identifies novel risk loci and implicates Abeta, Tau, immunity and lipid processing , 2018, bioRxiv.

[5]  I. Módy,et al.  Elevated TREM2 Gene Dosage Reprograms Microglia Responsivity and Ameliorates Pathological Phenotypes in Alzheimer’s Disease Models , 2018, Neuron.

[6]  M. Colonna,et al.  Humanized TREM2 mice reveal microglia-intrinsic and -extrinsic effects of R47H polymorphism , 2018, The Journal of experimental medicine.

[7]  M. Narici,et al.  Dysregulation of C-X-C motif ligand 10 during aging and association with cognitive performance , 2018, Neurobiology of Aging.

[8]  W. M. van der Flier,et al.  Genetic meta-analysis identifies 9 novel loci and functional pathways for Alzheimer’s disease risk , 2018, bioRxiv.

[9]  Kaur Alasoo,et al.  Shared genetic effects on chromatin and gene expression indicate a role for enhancer priming in immune response , 2018, Nature Genetics.

[10]  R. Marioni,et al.  GWAS on family history of Alzheimer’s disease , 2018, bioRxiv.

[11]  Thawfeek M. Varusai,et al.  The Reactome Pathway Knowledgebase , 2017, Nucleic acids research.

[12]  J. Schug,et al.  Integrated approach reveals diet, APOE genotype and sex affect immune response in APP mice. , 2018, Biochimica et biophysica acta. Molecular basis of disease.

[13]  T. Iwaki,et al.  Comparative profiling of cortical gene expression in Alzheimer’s disease patients and mouse models demonstrates a link between amyloidosis and neuroinflammation , 2017, Scientific Reports.

[14]  Nick C Fox,et al.  Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer's disease , 2017, Nature Genetics.

[15]  K. Hao,et al.  A common haplotype lowers PU.1 expression in myeloid cells and delays onset of Alzheimer's disease , 2017, Nature Neuroscience.

[16]  I. Amit,et al.  A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease , 2017, Cell.

[17]  W. Wurst,et al.  TREM2 deficiency impairs chemotaxis and microglial responses to neuronal injury , 2017, EMBO reports.

[18]  John Hardy,et al.  An additional k-means clustering step improves the biological features of WGCNA gene co-expression networks , 2017, BMC Systems Biology.

[19]  Rob Patro,et al.  Salmon provides fast and bias-aware quantification of transcript expression , 2017, Nature Methods.

[20]  Joseph K. Pickrell,et al.  Genetic regulatory effects modified by immune activation contribute to autoimmune disease associations , 2017, Nature Communications.

[21]  E. Hol,et al.  Transcriptional profiling of CD11c-positive microglia accumulating around amyloid plaques in a mouse model for Alzheimer's disease. , 2016, Biochimica et biophysica acta.

[22]  Ben A. Barres,et al.  Complement and microglia mediate early synapse loss in Alzheimer mouse models , 2016, Science.

[23]  Mariana Vargas-Caballero,et al.  Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer’s-like pathology , 2016, Brain : a journal of neurology.

[24]  Minoru Kanehisa,et al.  KEGG as a reference resource for gene and protein annotation , 2015, Nucleic Acids Res..

[25]  M. Robinson,et al.  Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. , 2015, F1000Research.

[26]  Gabor T. Marth,et al.  A global reference for human genetic variation , 2015, Nature.

[27]  Stein Aerts,et al.  i-cisTarget 2015 update: generalized cis-regulatory enrichment analysis in human, mouse and fly , 2015, Nucleic Acids Res..

[28]  D. Holtzman,et al.  TREM2 lipid sensing sustains microglia response in an Alzheimer’s disease model , 2015, Cell.

[29]  F. Edwards,et al.  A genome-wide gene-expression analysis and database in transgenic mice during development of amyloid or tau pathology. , 2015, Cell reports.

[30]  M. Heneka,et al.  CXCR3 promotes plaque formation and behavioral deficits in an Alzheimer's disease model. , 2015, The Journal of clinical investigation.

[31]  J. Satoh,et al.  A Comprehensive Profile of ChIP-Seq-Based PU.1/Spi1 Target Genes in Microglia , 2014, Gene regulation and systems biology.

[32]  A. Singleton,et al.  Genetic variability in the regulation of gene expression in ten regions of the human brain , 2014, Nature Neuroscience.

[33]  Nick C Fox,et al.  Gene-Wide Analysis Detects Two New Susceptibility Genes for Alzheimer's Disease , 2014, PLoS ONE.

[34]  C. Wallace,et al.  Bayesian Test for Colocalisation between Pairs of Genetic Association Studies Using Summary Statistics , 2013, PLoS genetics.

[35]  Nick C Fox,et al.  Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease , 2013, Nature Genetics.

[36]  Jennifer M. Pocock,et al.  Insights into TREM2 biology by network analysis of human brain gene expression data , 2013, Neurobiology of Aging.

[37]  Keith A. Johnson,et al.  CD33 Alzheimer’s disease locus: Altered monocyte function and amyloid biology , 2013, Nature Neuroscience.

[38]  Bradley T. Hyman,et al.  Alzheimer’s Disease Risk Gene CD33 Inhibits Microglial Uptake of Amyloid Beta , 2013, Neuron.

[39]  E. Hol,et al.  Cortical beta amyloid protein triggers an immune response, but no synaptic changes in the APPswe/PS1dE9 Alzheimer's disease mouse model , 2013, Neurobiology of Aging.

[40]  L. Tran,et al.  Integrated Systems Approach Identifies Genetic Nodes and Networks in Late-Onset Alzheimer’s Disease , 2013, Cell.

[41]  J. Donovan,et al.  Structural basis for cytosolic double-stranded RNA surveillance by human oligoadenylate synthetase 1 , 2013, Proceedings of the National Academy of Sciences.

[42]  A. Singleton,et al.  TREM2 variants in Alzheimer's disease. , 2013, The New England journal of medicine.

[43]  M. Heneka,et al.  NLRP3 is activated in Alzheimer´s disease and contributes to pathology in APP/PS1 mice , 2012, Nature.

[44]  J. Schneider,et al.  Overview and findings from the religious orders study. , 2012, Current Alzheimer research.

[45]  J. Schneider,et al.  Overview and findings from the rush Memory and Aging Project. , 2012, Current Alzheimer research.

[46]  Sampath Prahalad,et al.  Evaluation of genetic association between an ITGAM non-synonymous SNP (rs1143679) and multiple autoimmune diseases. , 2012, Autoimmunity reviews.

[47]  Seth Love,et al.  Genetic Evidence Implicates the Immune System and Cholesterol Metabolism in the Aetiology of Alzheimer's Disease , 2010, PloS one.

[48]  K. Elliott,et al.  Potential late-onset Alzheimer's disease-associated mutations in the ADAM10 gene attenuate {alpha}-secretase activity. , 2009, Human molecular genetics.

[49]  Steve Horvath,et al.  WGCNA: an R package for weighted correlation network analysis , 2008, BMC Bioinformatics.

[50]  B. Winblad,et al.  Decreased Fractalkine and Increased IP-10 Expression in Aged Brain of APPswe Transgenic Mice , 2008, Neurochemical Research.

[51]  John D. Storey,et al.  Capturing Heterogeneity in Gene Expression Studies by Surrogate Variable Analysis , 2007, PLoS genetics.

[52]  Hedi Peterson,et al.  g:Profiler—a web-based toolset for functional profiling of gene lists from large-scale experiments , 2007, Nucleic Acids Res..

[53]  V. Moskvina,et al.  Detailed Analysis of the Relative Power of Direct and Indirect Association Studies and the Implications for Their Interpretation , 2007, Human Heredity.

[54]  Cheng Li,et al.  Adjusting batch effects in microarray expression data using empirical Bayes methods. , 2007, Biostatistics.

[55]  S. Horvath,et al.  Conservation and evolution of gene coexpression networks in human and chimpanzee brains , 2006, Proceedings of the National Academy of Sciences.

[56]  S. Horvath,et al.  Analysis of oncogenic signaling networks in glioblastoma identifies ASPM as a molecular target , 2006, Proceedings of the National Academy of Sciences.

[57]  S. Horvath,et al.  Statistical Applications in Genetics and Molecular Biology , 2011 .

[58]  J. Hardy,et al.  The Amyloid Hypothesis of Alzheimer ’ s Disease : Progress and Problems on the Road to Therapeutics , 2009 .

[59]  C. Lemere,et al.  Inflammatory responses to amyloidosis in a transgenic mouse model of Alzheimer's disease. , 2001, The American journal of pathology.