Predictive Network Analysis Identifies JMJD6 and Other Novel Key Drivers in Alzheimer’s Disease

Despite decades of genetic studies on late onset Alzheimer’s disease (LOAD), the molecular mechanisms of Alzheimer’s disease (AD) remain unclear. Furthermore, different cell types in the central nervous system (CNS) play distinct roles in the onset and progression of AD pathology. To better comprehend the complex etiology of AD, we used an integrative approach to build robust predictive (causal) network models which were cross-validated over multiple large human multi-omics datasets in AD. We employed a published method to delineate bulk-tissue gene expression into single cell-type gene expression and integrated clinical and pathologic traits of AD, single nucleotide variation, and deconvoluted gene expression for the construction of predictive network models for each cell type in AD. With these predictive causal models, we are able to identify and prioritize robust key drivers of the AD-associated network state. In this study, we focused on neuron-specific network models and prioritized 19 predicted key drivers modulating AD pathology. These targets were validated via shRNA knockdown in human induced pluripotent stem cell (iPSC) derived neurons (iNs), in which 10 out of the 19 neuron-related targets (JMJD6, NSF, NUDT2, YWHAZ, RBM4, DCAF12, NDRG4, STXBP1, ATP1B1, and FIBP) significantly modulated levels of amyloid-beta and/or phosphorylated tau peptides in the postmitotic iNs. Most notably, knockdown of JMJD6 significantly altered the neurotoxic ratios of Aβ42 to 40 and p231-tau to total tau, indicating its potential therapeutic relevance to both amyloid and tau pathology in AD. Molecular validation by RNA sequencing (RNAseq) in iNs further confirmed the network structure, showing significant enrichment in differentially expressed genes after knockdown of the validated targets. Interestingly, our network model predicts that these 10 key drivers are upstream regulators of REST and VGF, two recently identified key regulators of AD pathogenesis.

[1]  V. Menon,et al.  Stem cell-derived neurons reflect features of protein networks, neuropathology, and cognitive outcome of their aged human donors , 2021, Neuron.

[2]  E. Schadt,et al.  Predictive network modeling in human induced pluripotent stem cells identifies key driver genes for insulin responsiveness , 2020, PLoS Comput. Biol..

[3]  Q. Dong,et al.  Genome-Wide Association Study of Brain Alzheimer's Disease-Related Metabolic Decline as Measured by [18F] FDG-PET Imaging. , 2020, Journal of Alzheimer's disease : JAD.

[4]  Bin Zhang,et al.  Multiscale causal networks identify VGF as a key regulator of Alzheimer’s disease , 2020, Nature Communications.

[5]  K. Setchell,et al.  Inhibition of Cdc42 activity extends lifespan and decreases circulating inflammatory cytokines in aged female C57BL/6 mice , 2020, Aging cell.

[6]  J. Peng,et al.  Optimal Nutrition Formulas for Patients Undergoing Surgery for Colorectal Cancer: A Bayesian Network Analysis , 2020, Nutrition and cancer.

[7]  R. Green,et al.  Genome‐wide association study of rate of cognitive decline in Alzheimer's disease patients identifies novel genes and pathways , 2020, Alzheimer's & dementia : the journal of the Alzheimer's Association.

[8]  D. Bennett,et al.  Single Cell-type Integrative Network Modeling Identified Novel Microglial-specific Targets for the Phagosome in Alzheimer’s disease , 2020, bioRxiv.

[9]  M. Aschner,et al.  The transcription factor REST up-regulates tyrosine hydroxylase and antiapoptotic genes and protects dopaminergic neurons against manganese toxicity , 2020, The Journal of Biological Chemistry.

[10]  B. Wang,et al.  BAP31 regulates IRAK1-dependent neuroinflammation in microglia , 2019, Journal of Neuroinflammation.

[11]  Kai Kang,et al.  CDSeq: A novel complete deconvolution method for dissecting heterogeneous samples using gene expression data , 2019, PLoS Comput. Biol..

[12]  Yen‐Hua Huang,et al.  Improved cell composition deconvolution method of bulk gene expression profiles to quantify subsets of immune cells , 2019, BMC Medical Genomics.

[13]  J. Bell,et al.  DNA Methylation Age and Physical and Cognitive Aging , 2019, The journals of gerontology. Series A, Biological sciences and medical sciences.

[14]  A. Ruiz,et al.  Genome-wide association analysis of dementia and its clinical endophenotypes reveal novel loci associated with Alzheimer's disease and three causality networks: The GR@ACE project , 2019, Alzheimer's & Dementia.

[15]  Fei Zou,et al.  SCDC: bulk gene expression deconvolution by multiple single-cell RNA sequencing references , 2019, bioRxiv.

[16]  B. Kerr,et al.  Writers and Readers of DNA Methylation/Hydroxymethylation in Physiological Aging and Its Impact on Cognitive Function , 2019, Neural plasticity.

[17]  R. Chang,et al.  The role of sleep deprivation and circadian rhythm disruption as risk factors of Alzheimer’s disease , 2019, Frontiers in Neuroendocrinology.

[18]  Nicole Rusk Expanded CIBERSORTx , 2019, Nature Methods.

[19]  Stefan Bonn,et al.  Deep-learning-based cell composition analysis from tissue expression profiles , 2019, bioRxiv.

[20]  Fabian J Theis,et al.  Current best practices in single‐cell RNA‐seq analysis: a tutorial , 2019, Molecular systems biology.

[21]  Manolis Kellis,et al.  Single-cell transcriptomic analysis of Alzheimer’s disease , 2019, Nature.

[22]  Ash A. Alizadeh,et al.  Determining cell-type abundance and expression from bulk tissues with digital cytometry , 2019, Nature Biotechnology.

[23]  Gary D Bader,et al.  Evaluation of methods to assign cell type labels to cell clusters from single-cell RNA-sequencing data , 2019, bioRxiv.

[24]  F. Feng,et al.  Efficacy and safety of targeted therapy for metastatic HER2-positive breast cancer in the first-line treatment: a Bayesian network meta-analysis , 2019, OncoTargets and therapy.

[25]  Gregory J. Hunt,et al.  Dtangle: Accurate and Robust Cell Type Deconvolution , 2018, Bioinform..

[26]  Charles C. White,et al.  Candidate‐based screening via gene modulation in human neurons and astrocytes implicates FERMT2 in A&bgr; and TAU proteostasis , 2018, Human molecular genetics.

[27]  Burkhard Tümmler,et al.  Faculty Opinions recommendation of A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. , 2018, Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature.

[28]  Cory C. Funk,et al.  Divergent brain gene expression patterns associate with distinct cell-specific tau neuropathology traits in progressive supranuclear palsy , 2018, Acta Neuropathologica.

[29]  Richard D. Smith,et al.  The human brainome: network analysis identifies HSPA2 as a novel Alzheimer’s disease target , 2018, Brain : a journal of neurology.

[30]  Aviv Regev,et al.  A revised airway epithelial hierarchy includes CFTR-expressing ionocytes , 2018, Nature.

[31]  K. Vakili,et al.  Hippo Signaling Pathway Dysregulation in Human Huntington’s Disease Brain and Neuronal Stem Cells , 2018, Scientific Reports.

[32]  David A Bennett,et al.  Religious Orders Study and Rush Memory and Aging Project. , 2018, Journal of Alzheimer's disease : JAD.

[33]  N. Mulder,et al.  Genome-Wide Association Study of Brain Connectivity Changes for Alzheimer’s Disease , 2018, Scientific Reports.

[34]  Allon M. Klein,et al.  The dynamics of gene expression in vertebrate embryogenesis at single-cell resolution , 2018, Science.

[35]  Fabian J Theis,et al.  Cell type atlas and lineage tree of a whole complex animal by single-cell transcriptomics , 2018, Science.

[36]  Lindsay A. Farrer,et al.  Genome-wide association study of Alzheimer's disease endophenotypes at prediagnosis stages , 2018, Alzheimer's & Dementia.

[37]  Klaus P. Ebmeier,et al.  Peripheral DNA methylation, cognitive decline and brain aging: pilot findings from the Whitehall II imaging study , 2018, Epigenomics.

[38]  Gregory J. Hunt,et al.  dtangle: accurate and fast cell-type deconvolution , 2018, bioRxiv.

[39]  Cory C. Funk,et al.  Conserved brain myelination networks are altered in Alzheimer's and other neurodegenerative diseases , 2018, Alzheimer's & Dementia.

[40]  R. Zukin,et al.  REST, a master transcriptional regulator in neurodegenerative disease , 2018, Current Opinion in Neurobiology.

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

[42]  K. Hao,et al.  A functional genomics predictive network model identifies regulators of inflammatory bowel disease , 2017, Nature Genetics.

[43]  A. Riva,et al.  DNA Methylation of Synaptic Genes in the Prefrontal Cortex Is Associated with Aging and Age-Related Cognitive Impairment , 2017, Front. Aging Neurosci..

[44]  J. C. Love,et al.  Cell-type Dependent Alzheimer's Disease Phenotypes: Probing the Biology of Selective Neuronal Vulnerability , 2017, Alzheimer's & Dementia.

[45]  Baptiste N. Jaeger,et al.  An environment-dependent transcriptional network specifies human microglia identity , 2017, Science.

[46]  Sarah A. Teichmann,et al.  Computational approaches for interpreting scRNA‐seq data , 2017, FEBS letters.

[47]  J. Haines,et al.  Genome-wide association study identifies four novel loci associated with Alzheimer’s endophenotypes and disease modifiers , 2017, Acta Neuropathologica.

[48]  Gaurav Pandey,et al.  Analysis of Transcriptional Variability in a Large Human iPSC Library Reveals Genetic and Non-genetic Determinants of Heterogeneity. , 2017, Cell stem cell.

[49]  D. Hume,et al.  Jmjd6, a JmjC Dioxygenase with Many Interaction Partners and Pleiotropic Functions , 2017, Front. Genet..

[50]  Göran Hallmans,et al.  Untangling the role of one-carbon metabolism in colorectal cancer risk: a comprehensive Bayesian network analysis , 2017, Scientific Reports.

[51]  Samuel L. Wolock,et al.  A Single-Cell Transcriptomic Map of the Human and Mouse Pancreas Reveals Inter- and Intra-cell Population Structure. , 2016, Cell systems.

[52]  James A. Eddy,et al.  Human whole genome genotype and transcriptome data for Alzheimer’s and other neurodegenerative diseases , 2016, Scientific Data.

[53]  Ralf Herwig,et al.  Analyzing and interpreting genome data at the network level with ConsensusPathDB , 2016, Nature Protocols.

[54]  Tom Michoel,et al.  Cardiometabolic risk loci share downstream cis- and trans-gene regulation across tissues and diseases , 2016, Science.

[55]  Fei Liu,et al.  Inference of Gene Regulatory Network Based on Local Bayesian Networks , 2016, PLoS Comput. Biol..

[56]  Trieu Nguyen,et al.  Integrative functional genomics identifies regulatory mechanisms at coronary artery disease loci , 2016, Nature Communications.

[57]  M. Ronaghi,et al.  Neuronal subtypes and diversity revealed by single-nucleus RNA sequencing of the human brain , 2016, Science.

[58]  G. Schellenberg,et al.  Gene expression, methylation and neuropathology correlations at progressive supranuclear palsy risk loci , 2016, Acta Neuropathologica.

[59]  J. Jia,et al.  An Overview of Genome-Wide Association Studies in Alzheimer’s Disease , 2016, Neuroscience Bulletin.

[60]  E. Chang,et al.  Purification and Characterization of Progenitor and Mature Human Astrocytes Reveals Transcriptional and Functional Differences with Mouse , 2016, Neuron.

[61]  Chuen-Mao Yang,et al.  NF-kappaB Signaling Pathways in Neurological Inflammation: A Mini Review , 2015, Front. Mol. Neurosci..

[62]  Yongshuai Jiang,et al.  Genome-wide haplotype association study identify TNFRSF1A, CASP7, LRP1B, CDH1 and TG genes associated with Alzheimer's disease in Caribbean Hispanic individuals , 2015, Oncotarget.

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

[64]  Shriprakash Sinha,et al.  Reproducibility of parameter learning with missing observations in naive Wnt Bayesian network trained on colorectal cancer samples and doxycycline-treated cell lines. , 2015, Molecular bioSystems.

[65]  Xiangru Xu DNA methylation and cognitive aging , 2015, Oncotarget.

[66]  S. Quake,et al.  A survey of human brain transcriptome diversity at the single cell level , 2015, Proceedings of the National Academy of Sciences.

[67]  Ash A. Alizadeh,et al.  Robust enumeration of cell subsets from tissue expression profiles , 2015, Nature Methods.

[68]  D. Holtzman,et al.  Sleep, circadian rhythms, and the pathogenesis of Alzheimer Disease , 2015, Experimental & Molecular Medicine.

[69]  G. von Heijne,et al.  Tissue-based map of the human proteome , 2015, Science.

[70]  Matthew E. Ritchie,et al.  limma powers differential expression analyses for RNA-sequencing and microarray studies , 2015, Nucleic acids research.

[71]  Eric E Schadt,et al.  A role for noncoding variation in schizophrenia. , 2014, Cell reports.

[72]  Rui Chang,et al.  Causal Inference in Biology Networks with Integrated Belief Propagation , 2014, Pacific Symposium on Biocomputing.

[73]  Carson C Chow,et al.  Second-generation PLINK: rising to the challenge of larger and richer datasets , 2014, GigaScience.

[74]  T. Maniatis,et al.  An RNA-Sequencing Transcriptome and Splicing Database of Glia, Neurons, and Vascular Cells of the Cerebral Cortex , 2014, The Journal of Neuroscience.

[75]  Margaret A. Pericak-Vance,et al.  Genome-Wide Association Meta-analysis of Neuropathologic Features of Alzheimer's Disease and Related Dementias , 2014, PLoS genetics.

[76]  Paul Theodor Pyl,et al.  HTSeq—a Python framework to work with high-throughput sequencing data , 2014, bioRxiv.

[77]  E. Davila,et al.  IL-1 Receptor-Associated Kinase Signaling and Its Role in Inflammation, Cancer Progression, and Therapy Resistance , 2014, Front. Immunol..

[78]  David A. Bennett,et al.  REST and Stress Resistance in Aging and Alzheimer’s Disease , 2014, Nature.

[79]  U. Oppermann,et al.  The roles of Jumonji-type oxygenases in human disease , 2014, Epigenomics.

[80]  Robert C. Green,et al.  Genome-wide association study of the rate of cognitive decline in Alzheimer's disease , 2014, Alzheimer's & Dementia.

[81]  A. Myers The Genetics of Gene Expression: Multiple Layers and Multiple Players , 2013 .

[82]  C. Reitz,et al.  Genome-wide Association Studies in Alzheimer’s Disease: A Review , 2013, Current Neurology and Neuroscience Reports.

[83]  Jun Zhu,et al.  Identification of Key Causal Regulators in Gene Networks , 2013 .

[84]  T. Südhof,et al.  Rapid Single-Step Induction of Functional Neurons from Human Pluripotent Stem Cells , 2013, Neuron.

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

[86]  Zhandong Liu,et al.  Digital sorting of complex tissues for cell type-specific gene expression profiles , 2013, BMC Bioinformatics.

[87]  A. Myers AD gene 3-D: moving past single layer genetic information to map novel loci involved in Alzheimer's disease. , 2012, Journal of Alzheimer's disease : JAD.

[88]  B. Yandell,et al.  Integrative Analysis of a Cross-Loci Regulation Network Identifies App as a Gene Regulating Insulin Secretion from Pancreatic Islets , 2012, PLoS genetics.

[89]  David Levine,et al.  A high-performance computing toolset for relatedness and principal component analysis of SNP data , 2012, Bioinform..

[90]  Ralf Herwig,et al.  The ConsensusPathDB interaction database: 2013 update , 2012, Nucleic Acids Res..

[91]  E. Pennacchietti,et al.  Glutamate decarboxylase‐dependent acid resistance in orally acquired bacteria: function, distribution and biomedical implications of the gadBC operon , 2012, Molecular microbiology.

[92]  Wei Li,et al.  RSeQC: quality control of RNA-seq experiments , 2012, Bioinform..

[93]  C. Korth,et al.  C-terminal fragment of N-cadherin accelerates synapse destabilization by amyloid-β. , 2012, Brain : a journal of neurology.

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

[95]  A. Myers The age of the “ome”: Genome, transcriptome and proteome data set collection and analysis , 2012, Brain Research Bulletin.

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

[97]  Chris Williams,et al.  RNA-SeQC: RNA-seq metrics for quality control and process optimization , 2012, Bioinform..

[98]  K. Hao,et al.  Bayesian method to predict individual SNP genotypes from gene expression data , 2012, Nature Genetics.

[99]  Rachel B. Brem,et al.  Stitching together Multiple Data Dimensions Reveals Interacting Metabolomic and Transcriptomic Networks That Modulate Cell Regulation , 2012, PLoS biology.

[100]  K. Hansen,et al.  Removing technical variability in RNA-seq data using conditional quantile normalization , 2012, Biostatistics.

[101]  R. Faull,et al.  Population-specific expression analysis (PSEA) reveals molecular changes in diseased brain , 2011, Nature Methods.

[102]  A. Kasarskis,et al.  Identification of causal genes, networks, and transcriptional regulators of REM sleep and wake. , 2011, Sleep.

[103]  J. David Sweatt,et al.  Epigenetic Mechanisms in Cognition , 2011, Neuron.

[104]  Andrey A. Shabalin,et al.  Matrix eQTL: ultra fast eQTL analysis via large matrix operations , 2011, Bioinform..

[105]  Joseph O Deasy,et al.  A Bayesian network approach for modeling local failure in lung cancer , 2011, Physics in medicine and biology.

[106]  S. Shimohama,et al.  N-cadherin Regulates p38 MAPK Signaling via Association with JNK-associated Leucine Zipper Protein , 2010, The Journal of Biological Chemistry.

[107]  Ralf Herwig,et al.  ConsensusPathDB: toward a more complete picture of cell biology , 2010, Nucleic Acids Res..

[108]  Yuhai Zhao,et al.  Differential Regulation of Interleukin-1 Receptor-associated Kinase-1 (IRAK-1) and IRAK-2 by MicroRNA-146a and NF-κB in Stressed Human Astroglial Cells and in Alzheimer Disease* , 2010, The Journal of Biological Chemistry.

[109]  Trey Ideker,et al.  Evidence mining and novelty assessment of protein–protein interactions with the ConsensusPathDB plugin for Cytoscape , 2010, Bioinform..

[110]  Dario Strbenac,et al.  Repitools: an R package for the analysis of enrichment-based epigenomic data , 2010, Bioinform..

[111]  Xia Yang,et al.  Liver and Adipose Expression Associated SNPs Are Enriched for Association to Type 2 Diabetes , 2010, PLoS genetics.

[112]  Xia Yang,et al.  Integrating pathway analysis and genetics of gene expression for genome-wide association studies. , 2010, American journal of human genetics.

[113]  M. Robinson,et al.  A scaling normalization method for differential expression analysis of RNA-seq data , 2010, Genome Biology.

[114]  Wei Zhang,et al.  A Bayesian Partition Method for Detecting Pleiotropic and Epistatic eQTL Modules , 2010, PLoS Comput. Biol..

[115]  Mark D. Robinson,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[116]  Z. Modrušan,et al.  Deconvolution of Blood Microarray Data Identifies Cellular Activation Patterns in Systemic Lupus Erythematosus , 2009, PloS one.

[117]  Gonçalo R. Abecasis,et al.  The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..

[118]  P. Donnelly,et al.  A Flexible and Accurate Genotype Imputation Method for the Next Generation of Genome-Wide Association Studies , 2009, PLoS genetics.

[119]  Eric E Schadt,et al.  Multi-tissue coexpression networks reveal unexpected subnetworks associated with disease. , 2009 .

[120]  D. Stephan,et al.  Genetic control of human brain transcript expression in Alzheimer disease. , 2009, American journal of human genetics.

[121]  Lior Pachter,et al.  Sequence Analysis , 2020, Definitions.

[122]  Cole Trapnell,et al.  Ultrafast and memory-efficient alignment of short DNA sequences to the human genome , 2009, Genome Biology.

[123]  Ralf Herwig,et al.  ConsensusPathDB—a database for integrating human functional interaction networks , 2008, Nucleic Acids Res..

[124]  Rachel B. Brem,et al.  Integrating large-scale functional genomic data to dissect the complexity of yeast regulatory networks , 2008, Nature Genetics.

[125]  H. Stefánsson,et al.  Genetics of gene expression and its effect on disease , 2008, Nature.

[126]  O. Combarros,et al.  14-3-3 Zeta and Tau Genes Interactively Decrease Alzheimer’s Disease Risk , 2008, Dementia and Geriatric Cognitive Disorders.

[127]  D. Stephan,et al.  A survey of genetic human cortical gene expression , 2007, Nature Genetics.

[128]  M. Owen,et al.  Genome-wide linkage analysis of 723 affected relative pairs with late-onset Alzheimer's disease. , 2007, Human molecular genetics.

[129]  Anatol C. Kreitzer,et al.  Aberrant Excitatory Neuronal Activity and Compensatory Remodeling of Inhibitory Hippocampal Circuits in Mouse Models of Alzheimer's Disease , 2007, Neuron.

[130]  Manuel A. R. Ferreira,et al.  PLINK: a tool set for whole-genome association and population-based linkage analyses. , 2007, American journal of human genetics.

[131]  Jun Zhu,et al.  Increasing the Power to Detect Causal Associations by Combining Genotypic and Expression Data in Segregating Populations , 2007, PLoS Comput. Biol..

[132]  D. Reich,et al.  Population Structure and Eigenanalysis , 2006, PLoS genetics.

[133]  A. Arnold,et al.  Tissue-specific expression and regulation of sexually dimorphic genes in mice. , 2006, Genome research.

[134]  S. Horvath,et al.  Evidence for anti-Burkitt tumour globulins in Burkitt tumour patients and healthy individuals. , 1967, British Journal of Cancer.

[135]  E E Schadt,et al.  Integrating genotypic and expression data in a segregating mouse population to identify 5-lipoxygenase as a susceptibility gene for obesity and bone traits , 2005, Nature Genetics.

[136]  J. Castle,et al.  An integrative genomics approach to infer causal associations between gene expression and disease , 2005, Nature Genetics.

[137]  Eric E Schadt,et al.  Cis-acting expression quantitative trait loci in mice. , 2005, Genome research.

[138]  A. Delacourte,et al.  Examination of stress-related genes in human temporal versus occipital cortex in the course of neurodegeneration: involvement of 14-3-3 zeta in this dynamic process , 2004, Neuroscience Letters.

[139]  T. Iwamoto,et al.  14-3-3 proteins and zeta isoform containing neurofibrillary tangles in patients with Alzheimer’s disease , 2004, Acta Neuropathologica.

[140]  J. Zhu,et al.  An integrative genomics approach to the reconstruction of gene networks in segregating populations , 2004, Cytogenetic and Genome Research.

[141]  J. Ávila,et al.  Zeta 14-3-3 protein favours the formation of human tau fibrillar polymers , 2004, Neuroscience Letters.

[142]  R. Stoughton,et al.  Genetics of gene expression surveyed in maize, mouse and man , 2003, Nature.

[143]  H. Braak,et al.  Staging of brain pathology related to sporadic Parkinson’s disease , 2003, Neurobiology of Aging.

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

[145]  Y. Benjamini,et al.  Controlling the false discovery rate in behavior genetics research , 2001, Behavioural Brain Research.

[146]  B. Crain,et al.  Interlaboratory Comparison of Neuropathology Assessments in Alzheimer's Disease: A Study of the Consortium to Establish a Registry for Alzheimer's Disease (CERAD) , 1994, Journal of neuropathology and experimental neurology.

[147]  S. M. Sumi,et al.  The Consortium to Establish a Registry for Alzheimer's Disease (CERAD) , 1991, Neurology.

[148]  Robert E. Tarjan,et al.  Depth-First Search and Linear Graph Algorithms , 1972, SIAM J. Comput..

[149]  Y. Lee,et al.  Genome-wide pathway analysis of a genome-wide association study on Alzheimer’s disease , 2014, Neurological Sciences.

[150]  N. Bresolin,et al.  Genetics and expression analysis of the specificity protein 4 gene (SP4) in patients with Alzheimer's disease and frontotemporal lobar degeneration. , 2012, Journal of Alzheimer's Disease.

[151]  M. Murphy,et al.  Alzheimer's disease and the amyloid-beta peptide. , 2010, Journal of Alzheimer's disease : JAD.

[152]  Lakhmi C. Jain,et al.  Introduction to Bayesian Networks , 2008 .

[153]  H. Braak,et al.  Neuropathological stageing of Alzheimer-related changes , 2004, Acta Neuropathologica.

[154]  N. Holbrook,et al.  Oxidative stress-induced phospholipase C-gamma 1 activation enhances cell survival. , 2001, The Journal of biological chemistry.

[155]  N. Seyfried,et al.  Convergence of independent DISC 1 mutations on impaired neurite growth via decreased UNC 5 D expression , 2022 .