A review on omics-based biomarkers discovery for Alzheimer's disease from the bioinformatics perspectives: Statistical approach vs machine learning approach
Tài liệu tham khảo
2007, Dementia: a NICE-SCIE guideline on supporting people with dementia and their carers in health and social care. National clinical practice guideline number 42, Br. Psychol.Soc.
Mucke, 2009, Alzheimer's disease, Nature, 461, 895, 10.1038/461895a
Wolk, 2016
Swerdlow, 2007, Is aging part of Alzheimer's disease, or is Alzheimer's disease part of aging?, Neurobiol. Aging, 28, 1465, 10.1016/j.neurobiolaging.2006.06.021
Prince, 2015, World alzheimer report 2015-the global impact of dementia: an analysis of prevalence, incidence, cost and trends, Alzheimer's Dis. Int., 84
Alzheimer’s Association, 2020, Alzheimer's disease facts and figures, Alzheimers, Dementia, 16, 391
Craig, 2008, Complex diseases: research and applications, Nat. Educ., 1, 184
Mitchell, 2012, What is complex about complex disorders?, Genome Biol., 13, 1, 10.1186/gb-2012-13-1-237
Micheel, 2012, Omics-based Clinical Discovery: Science, Technology, and Applications
Vailati-Riboni, 2017, What Are Omics Sciences?, 1
Awada, 2015, Early and late-onset Alzheimer's disease: what are the differences?, J. Neurosci. Rural Pract., 6, 455, 10.4103/0976-3147.154581
Bekris, 2010, Genetics of alzheimer disease, J. Geriatr. Psychiatr. Neurol., 23, 213, 10.1177/0891988710383571
Jansen, 2019, Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer's disease risk, Nat. Genet., 51, 404, 10.1038/s41588-018-0311-9
Wightman, 2020
Hardy, 2002, The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics, Science, 297, 353, 10.1126/science.1072994
Yan, 2019, Integrated identification of key genes and pathways in Alzheimer's disease via comprehensive bioinformatical analyses, Hereditas, 156, 1, 10.1186/s41065-019-0101-0
Jung, 2018, Genomics: new light on Alzheimer's disease research, Int. J. Mol. Sci., 19, 3771, 10.3390/ijms19123771
Calabrò, 2021, The biological pathways of Alzheimer disease: a review, AIMS Neurosci, 8, 86, 10.3934/Neuroscience.2021005
Sancesario, 2018, Alzheimer's disease in the omics era, Clin. Biochem., 59, 9, 10.1016/j.clinbiochem.2018.06.011
Li, 2018, Heterogeneity analysis and diagnosis of complex diseases based on deep learning method, Sci. Rep., 8, 1
Morris, 2017, Statistical contributions to bioinformatics: design, modelling, structure learning and integration, Stat. Model. Int. J., 17, 245, 10.1177/1471082X17698255
Vasaikar, 2018, LinkedOmics: analyzing multi-omics data within and across 32 cancer types, Nucleic Acids Res., 46, 956, 10.1093/nar/gkx1090
Lockhart, 2000, Genomics, gene expression and DNA arrays, Nature, 405, 827, 10.1038/35015701
Belgard, 2011, A transcriptomic atlas of mouse neocortical layers, Neuron, 71, 605, 10.1016/j.neuron.2011.06.039
Mercer, 2008, Specific expression of long noncoding RNAs in the mouse brain, Proc. Natl. Acad. Sci. Unit. States Am., 105, 716, 10.1073/pnas.0706729105
Wu, 2020, Functional roles and networks of non-coding RNAs in the pathogenesis of neurodegenerative diseases, J. Biomed. Sci., 27, 1, 10.1186/s12929-020-00636-z
Wang, 2019, MicroRNAs in Alzheimer's disease, Front. Genet., 10, 153, 10.3389/fgene.2019.00153
Baek, 2008, The impact of microRNAs on protein output, Nature, 455, 64, 10.1038/nature07242
Lei, 2019, Analyzing the regulation of miRNAs on protein-protein interaction network in Hodgkin lymphoma, BMC Bioinf., 20, 1, 10.1186/s12859-019-3041-9
Choi, 2004, Oxidative modifications and down-regulation of ubiquitin carboxyl-terminal hydrolase L1 associated with idiopathic Parkinson's and Alzheimer's diseases, J. Biol. Chem., 279, 13256, 10.1074/jbc.M314124200
Choi, 2005, Oxidative modifications and aggregation of Cu, Zn-superoxide dismutase associated with Alzheimer and Parkinson diseases, J. Biol. Chem., 280, 11648, 10.1074/jbc.M414327200
Choi, 2006, Oxidative damage of DJ-1 is linked to sporadic Parkinson and Alzheimer diseases, J. Biol. Chem., 281, 10816, 10.1074/jbc.M509079200
Schonberger, 2001, Proteomic analysis of the brain in Alzheimer's disease: molecular phenotype of a complex disease process, Proteom. Int. Educ., 1, 1519
Korolainen, 2010, An update on clinical proteomics in Alzheimer's research, J. Neurochem., 112, 1386, 10.1111/j.1471-4159.2009.06558.x
Zhang, 2018, Integrated proteomics and network analysis identifies protein hubs and network alterations in Alzheimer's disease, Acta Neuropathologica Commun, 6, 1, 10.1186/s40478-018-0524-2
Johnson, 2018, Deep proteomic network analysis of Alzheimer's disease brain reveals alterations in RNA binding proteins and RNA splicing associated with disease, Mol. Neurodegener., 13, 1, 10.1186/s13024-018-0282-4
Higginbotham, 2019, Network analysis of a membrane-enriched brain proteome across stages of Alzheimer's disease, Proteomes, 7, 30, 10.3390/proteomes7030030
Dai, 2018, Effects of APOE genotype on brain proteomic network and cell type changes in Alzheimer's disease, Front. Mol. Neurosci., 11, 454, 10.3389/fnmol.2018.00454
Schubert, 2018, Targeted proteomic analysis of cognitive dysfunction in remitted major depressive disorder: opportunities of multi-omics approaches towards predictive, preventive, and personalized psychiatry, J. Proteom., 188, 63, 10.1016/j.jprot.2018.02.023
Yerlekar, 2014, A review on study and comparison between 2D gel electrophoresis and mass spectrometry, IOSR J. Comput. Eng., 16, 97, 10.9790/0661-162597104
Encarnación, 2005, Comparative proteomics using 2-D gel electrophoresis and mass spectrometry as tools to dissect stimulons and regulons in bacteria with sequenced or partially sequenced genomes, Biol. Proced. Online, 7, 117, 10.1251/bpo110
Ramazi, 2021, Posttranslational modifications in proteins: resources, tools and prediction methods, Database, 2021
Ramesh, 2020, Role of post‐translational modifications in Alzheimer's disease, Chembiochem, 21, 1052, 10.1002/cbic.201900573
Marcelli, 2018, The involvement of post-translational modifications in Alzheimer's disease, Curr. Alzheimer Res., 15, 313, 10.2174/1567205014666170505095109
Liu, 2011, Detecting and analyzing differentially activated pathways in brain regions of Alzheimer's disease patients, Mol. Biosyst., 7, 1441, 10.1039/c0mb00325e
Lamichhane, 2018, An overview of metabolomics data analysis: current tools and future perspectives, Compr. Anal. Chem., 82, 387, 10.1016/bs.coac.2018.07.001
Nishiumi, 2014, Metabolomics for biomarker discovery in gastroenterological cancer, Metabolites, 4, 547, 10.3390/metabo4030547
Hunsberger, 2020, Divergence in the metabolome between natural aging and Alzheimer's disease, Sci. Rep., 10, 1
Wilkins, 2018, Application of metabolomics in Alzheimer's disease, Front. Neurol., 8, 719, 10.3389/fneur.2017.00719
Cohen, 2009, Basal cerebral metabolism may modulate the cognitive effects of Aβ in mild cognitive impairment: an example of brain reserve, J. Neurosci., 29, 14770, 10.1523/JNEUROSCI.3669-09.2009
Pagani, 2017, Early identification of MCI converting to AD: a FDG PET study, Eur. J. Nucl. Med. Mol. Imag., 44, 2042, 10.1007/s00259-017-3761-x
Shah, 2015, Integrated metabolomics and genomics: systems approaches to biomarkers and mechanisms of cardiovascular disease, Circulation: Cardiovasc. Genet., 8, 410
Li, 2019, Statistically correlating NMR spectra and LC-MS data to facilitate the identification of individual metabolites in metabolomics mixtures, Anal. Bioanal. Chem., 411, 1301, 10.1007/s00216-019-01600-z
Sayers, 2020, Database resources of the national center for biotechnology information, Nucleic Acids Res., 48, 9, 10.1093/nar/gkz899
Mary-Huard, 2006, Introduction to statistical methods for microarray data analysis, Math. Comput. Methods Biol. Paris: Hermann
Garson, 2012
Nahm, 2016, Nonparametric statistical tests for the continuous data: the basic concept and the practical use, Korean J. Anesthesiol., 69, 8, 10.4097/kjae.2016.69.1.8
Gao, 2005, Nonparametric tests for differential gene expression and interaction effects in multi-factorial microarray experiments, BMC Bioinf., 6, 1, 10.1186/1471-2105-6-186
Mao, 2015, Association of CD33 and MS4A cluster variants with Alzheimer's disease in East Asian populations, Neurosci. Lett., 609, 235, 10.1016/j.neulet.2015.10.007
Doyle, 2014, Predicting progression of Alzheimer's disease using ordinal regression, PLoS One, 9, 10.1371/journal.pone.0105542
Teipel, 2015, The relative importance of imaging markers for the prediction of Alzheimer's disease dementia in mild cognitive impairment—beyond classical regression, Neuroimage Clin, 8, 583, 10.1016/j.nicl.2015.05.006
Tabarestani, 2018, Profile-Specific Regression Model for Progression Prediction of Alzheimer's Disease Using Longitudinal Data, 1353
Xiao, 2021, Early diagnosis model of Alzheimer's disease based on sparse logistic regression with the generalized elastic net, Biomed. Signal Process Control, 66, 10.1016/j.bspc.2020.102362
Zhou, 2020, Genetic and polygenic risk score analysis for Alzheimer's disease in the Chinese population, Alzheimers. Dement., 12
Seo, 2016, RNA-seq analysis for detecting quantitative trait-associated genes, Sci. Rep., 6, 1, 10.1038/srep24375
Lee, 2020, Prediction of Alzheimer's disease using blood gene expression data, Sci. Rep., 10, 1
Booij, 2011, A gene expression pattern in blood for the early detection of Alzheimer's disease, J. Alzheim. Dis., 23, 109, 10.3233/JAD-2010-101518
Li, 2018, Systematic analysis and biomarker study for Alzheimer's disease, Sci. Rep., 8, 1, 10.1038/s41598-018-35789-3
Chen, 2020, Spatial transcriptomics and in situ sequencing to study Alzheimer's disease, Cell, 182, 976, 10.1016/j.cell.2020.06.038
Tan, 2021, Associations of Alzheimer's disease risk variants with gene expression, amyloidosis, tauopathy, and neurodegeneration, Alzheimer's Res. Ther., 13, 1, 10.1186/s13195-020-00755-7
Marques-Coelho, 2021, Differential transcript usage unravels gene expression alterations in Alzheimer's disease human brains, npj Aging Mech. Dis., 7, 1, 10.1038/s41514-020-00052-5
Alexiou, 2017, A Bayesian model for the prediction and early diagnosis of Alzheimer's disease, Front. Aging Neurosci., 9, 77, 10.3389/fnagi.2017.00077
Zhang, 2013, Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer's disease, Cell, 153, 707, 10.1016/j.cell.2013.03.030
Jiménez-Jacinto, 2019, Integrative differential expression analysis for multiple experiments (IDEAMEX): a web server tool for integrated rna-seq data analysis, Front. Genet., 10, 279, 10.3389/fgene.2019.00279
Ihaka, 1996, R: a language for data analysis and graphics, J. Comput. Graph Stat., 5, 299
Espuny-Camacho, 2017, Hallmarks of Alzheimer's disease in stem-cell-derived human neurons transplanted into mouse brain, Neuron, 93, 1066, 10.1016/j.neuron.2017.02.001
Magistri, 2015, Transcriptomics profiling of Alzheimer's disease reveal neurovascular defects, altered amyloid-β homeostasis, and deregulated expression of long noncoding RNAs, J. Alzheim. Dis., 48, 647, 10.3233/JAD-150398
Sekar, 2015, Alzheimer's disease is associated with altered expression of genes involved in immune response and mitochondrial processes in astrocytes, Neurobiol. Aging, 36, 583, 10.1016/j.neurobiolaging.2014.09.027
Currais, 2015, A comprehensive multiomics approach toward understanding the relationship between aging and dementia, Aging (Albany NY), 7, 937, 10.18632/aging.100838
Zhou, 2015, Identification of Alzheimer's disease–associated long noncoding RNAs, Neurobiol. Aging, 36, 2925, 10.1016/j.neurobiolaging.2015.07.015
Carrasquillo, 2011, Replication of BIN1 association with Alzheimer's disease and evaluation of genetic interactions, J. Alzheim. Dis., 24, 751, 10.3233/JAD-2011-101932
Belbin, 2011, Investigation of 15 of the top candidate genes for late-onset Alzheimer's disease, Hum. Genet., 129, 273, 10.1007/s00439-010-0924-2
Skillbäck, 2013, Cerebrospinal fluid biomarkers for Alzheimer disease and subcortical axonal damage in 5,542 clinical samples, Alzheimer's Res. Ther., 5, 1, 10.1186/alzrt212
Müller, 2014, MicroRNAs in Alzheimer's disease: differential expression in hippocampus and cell-free cerebrospinal fluid, Neurobiol. Aging, 35, 152, 10.1016/j.neurobiolaging.2013.07.005
Li, 2017, Identification of molecular alterations in leukocytes from gene expression profiles of peripheral whole blood of Alzheimer's disease, Sci. Rep., 7, 1
Chang, 2019, APOE-MS4A genetic interactions are associated with executive dysfunction and network abnormality in clinically mild Alzheimer's disease, Neuroimage Clin, 21, 101621, 10.1016/j.nicl.2018.101621
Sathe, 2019, Quantitative proteomic profiling of cerebrospinal fluid to identify candidate biomarkers for Alzheimer's disease, Proteonomics Clin. Appl., 13
Kao, 2017, Pathway analysis of complex diseases for GWAS, extending to consider rare variants, multi-omics and interactions, Biochim. Biophys. Acta Gen. Subj., 1861, 335, 10.1016/j.bbagen.2016.11.030
Bush, 2012, Chapter 11: genome-wide association studies, PLoS Comput. Biol., 8, 10.1371/journal.pcbi.1002822
Cuyvers, 2016, Genetic variations underlying Alzheimer's disease: evidence from genome-wide association studies and beyond, Lancet Neurol., 15, 857, 10.1016/S1474-4422(16)00127-7
Harold, 2009, Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer's disease, Nat. Genet., 41, 1088, 10.1038/ng.440
Hollingworth, 2011, Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer's disease, Nat. Genet., 43, 429, 10.1038/ng.803
Seshadri, 2010, Genome-wide analysis of genetic loci associated with Alzheimer disease, Jama, 303, 1832, 10.1001/jama.2010.574
Lambert, 2009, Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer's disease, Nat. Genet., 41, 1094, 10.1038/ng.439
Naj, 2011, Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer's disease, Nat. Genet., 43, 436, 10.1038/ng.801
Beecham, 2009, Genome-wide association study implicates a chromosome 12 risk locus for late-onset Alzheimer disease, Am. J. Hum. Genet., 84, 35, 10.1016/j.ajhg.2008.12.008
Grupe, 2007, Evidence for novel susceptibility genes for late-onset Alzheimer's disease from a genome-wide association study of putative functional variants, Hum. Mol. Genet., 16, 865, 10.1093/hmg/ddm031
Li, 2008, Candidate single-nucleotide polymorphisms from a genomewide association study of Alzheimer disease, Arch. Neurol., 65, 45, 10.1001/archneurol.2007.3
Abraham, 2008, A genome-wide association study for late-onset Alzheimer's disease using DNA pooling, BMC Med. Genom., 1, 1, 10.1186/1755-8794-1-44
Hohman, 2016, Discovery of gene-gene interactions across multiple independent data sets of late onset Alzheimer disease from the Alzheimer Disease Genetics Consortium, Neurobiol. Aging, 38, 141, 10.1016/j.neurobiolaging.2015.10.031
Lambert, 2013, Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease, Nat. Genet., 45, 1452, 10.1038/ng.2802
Harris, 2018, GWAS on family history of Alzheimer's disease, Transl. Psychiatry, 8.1, 1
Porcu, 2019, Mendelian randomization integrating GWAS and eQTL data reveals genetic determinants of complex and clinical traits, Nat. Commun., 10, 1, 10.1038/s41467-019-10936-0
Ma, 2020, Alzheimer's disease GWAS weighted by multi‐omics and endophenotypes identifies novel risk loci: genetics/genetic factors of Alzheimer's disease, Alzheimers. Dement., 16, 10.1002/alz.043977
Warnat, 2005, Cross-platform analysis of cancer microarray data improves gene expression based classification of phenotypes, BMC Bioinf., 6, 1, 10.1186/1471-2105-6-265
Templin, 2002, Protein microarray technology, drug discov, Today Off., 7, 815
Tan, 2018, Integrative machine learning analysis of multiple gene expression profiles in cervical cancer, PeerJ, 6, e5285, 10.7717/peerj.5285
Jun, 2010, Meta-analysis confirms CR1, CLU, and PICALM as alzheimer disease risk loci and reveals interactions with APOE genotypes, Arch. Neurol., 67, 1473, 10.1001/archneurol.2010.201
Patel, 2019, A meta-analysis of Alzheimer's disease brain transcriptomic data, J. Alzheim. Dis., 68, 1635, 10.3233/JAD-181085
Su, 2019, Meta-analysis of gene expression and identification of biological regulatory mechanisms in Alzheimer's disease, Front. Neurosci., 13, 633, 10.3389/fnins.2019.00633
Potkin, 2009, Hippocampal atrophy as a quantitative trait in a genome-wide association study identifying novel susceptibility genes for Alzheimer's disease, PLoS One, 4, 10.1371/journal.pone.0006501
Ping, 2018, Global quantitative analysis of the human brain proteome in Alzheimer's and Parkinson's Disease, Sci. Data, 5, 1, 10.1038/sdata.2018.36
Keck, 2017, A systems level analysis of epileptogenesis-associated proteome alterations, Neurobiol. Dis., 105, 164, 10.1016/j.nbd.2017.05.017
Miller, 2008, A systems level analysis of transcriptional changes in Alzheimer's disease and normal aging, J. Neurosci., 28, 1410, 10.1523/JNEUROSCI.4098-07.2008
Seyfried, 2017, A multi-network approach identifies protein-specific co-expression in asymptomatic and symptomatic Alzheimer's disease, Cell Syst, 4, 60, 10.1016/j.cels.2016.11.006
Ashburner, 2000, Gene ontology: tool for the unification of biology, Nat. Genet., 25, 25, 10.1038/75556
Mitrea, 2013, Methods and approaches in the topology-based analysis of biological pathways, Front. Physiol., 4, 278, 10.3389/fphys.2013.00278
García-Campos, 2015, Pathway analysis: state of the art, Front. Physiol., 6, 383, 10.3389/fphys.2015.00383
Twine, 2011, Whole transcriptome sequencing reveals gene expression and splicing differences in brain regions affected by Alzheimer's disease, PLoS One, 6, 10.1371/journal.pone.0016266
Subramanian, 2005, Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles, Proc. Natl. Acad. Sci. Unit. States Am., 102, 15545, 10.1073/pnas.0506580102
Moradifard, 2018, Analysis of microRNA and gene expression profiles in Alzheimer's disease: a meta-analysis approach, Sci. Rep., 8, 1, 10.1038/s41598-018-20959-0
Kanehisa, 2000, KEGG: kyoto encyclopedia of genes and genomes, Nucleic Acids Res., 28, 27, 10.1093/nar/28.1.27
Harris, 2004, The Gene Ontology (GO) database and informatics resource, Nucleic Acids Res., 32, 258, 10.1093/nar/gkh036
Fabregat, 2018, The reactome pathway knowledgebase, Nucleic Acids Res., 46, 649, 10.1093/nar/gkx1132
Pico, 2008, WikiPathways: pathway editing for the people, PLoS Biol., 6, e184, 10.1371/journal.pbio.0060184
Mizuno, 2012, AlzPathway: a comprehensive map of signaling pathways of Alzheimer's disease, BMC Syst. Biol., 6, 1, 10.1186/1752-0509-6-52
Scuderi, 2020, Neuroinflammation in Alzheimer's disease: friend or foe?, Faseb. J., 34, 10.1096/fasebj.2020.34.s1.00381
Delgado, 2021, Neuroinflammation in Alzheimer's disease and beneficial action of luteolin, Biofactors, 47, 207, 10.1002/biof.1714
Cioffi, 2019, Molecular mechanisms and genetics of oxidative stress in Alzheimer's disease, J. Alzheim. Dis., 72, 981, 10.3233/JAD-190863
Wang, 2020, Mitochondria dysfunction in the pathogenesis of Alzheimer's disease: recent advances, Mol. Neurodegener., 15, 1, 10.1186/s13024-020-00376-6
Bell, 2021, Mitochondrial dysfunction in Alzheimer's disease: a biomarker of the future?, Biomedica, 9, 63, 10.3390/biomedicines9010063
Loera-Valencia, 2019, Alterations in cholesterol metabolism as a risk factor for developing Alzheimer's disease: potential novel targets for treatment, J. Steroid Biochem. Mol. Biol., 190, 104, 10.1016/j.jsbmb.2019.03.003
Sun, 2019, The application of weighted gene co-expression network analysis in identifying key modules and hub genes associated with disease status in Alzheimer's disease, Ann. Transl. Med., 7, 800, 10.21037/atm.2019.12.59
Samuel, 1959, Some studies in machine learning using the game of checkers, IBM J. Res. Dev., 3, 210, 10.1147/rd.33.0210
Provost, 1998, Glossary of terms, J. Mach. Learn., 30, 271, 10.1023/A:1007442505281
Faizal, 2021, A review of risk prediction models in cardiovascular disease: conventional approach vs. artificial intelligent approach, Comput. Methods Progr. Biomed.
Xu, 2019, 1
Madhamshettiwar, 2012, Gene regulatory network inference: evaluation and application to ovarian cancer allows the prioritization of drug targets, Genome Med., 4, 1, 10.1186/gm340
Reus, 2020, Alzheimer's Disease Neuroimaging Initiative, Degree of genetic liability for Alzheimer's disease associated with specific proteomic profiles in cerebrospinal fluid, Neurobiol. Aging, 93, 10.1016/j.neurobiolaging.2020.03.012
Toschi, 2019, Biomarker-guided clustering of Alzheimer's disease clinical syndromes, Neurobiol. Aging, 83, 42, 10.1016/j.neurobiolaging.2019.08.032
Alashwal, 2019, The application of unsupervised clustering methods to Alzheimer's disease, Front. Comput. Neurosci., 13, 31, 10.3389/fncom.2019.00031
Sudduth, 2013, Neuroinflammatory phenotype in early Alzheimer's disease, Neurobiol. Aging, 34, 1051, 10.1016/j.neurobiolaging.2012.09.012
Wallin, 2010, CSF biomarkers predict a more malignant outcome in Alzheimer disease, Neurology, 74, 1531, 10.1212/WNL.0b013e3181dd4dd8
Agarwal, 2015, Identification of a small set of plasma signalling proteins using neural network for prediction of Alzheimer's disease, Bioinformatics, 31, 2505, 10.1093/bioinformatics/btv173
Schrider, 2018, Supervised machine learning for population genetics: a new paradigm, Trends Genet., 34, 301, 10.1016/j.tig.2017.12.005
Xu, 2018, An efficient classifier for Alzheimer's disease genes identification, Molecules, 23, 3140, 10.3390/molecules23123140
Huang, 2018, Revealing Alzheimer's disease genes spectrum in the whole-genome by machine learning, BMC Neurol., 18, 1, 10.1186/s12883-017-1010-3
Lugli, 2015, Plasma exosomal miRNAs in persons with and without Alzheimer disease: altered expression and prospects for biomarkers, PLoS One, 10, 10.1371/journal.pone.0139233
Zhou, 2019, Analysis of long noncoding RNAs highlights region-specific altered expression patterns and diagnostic roles in Alzheimer's disease, Briefings Bioinf., 20, 598, 10.1093/bib/bby021
Long, 2016, Discovery of novel biomarkers for Alzheimer's disease from blood, Dis. Markers, 10.1155/2016/4250480
Fehlbaum-Beurdeley, 2010, Toward an Alzheimer's disease diagnosis via high-resolution blood gene expression, Alzheimers. Dement., 6, 25, 10.1016/j.jalz.2009.07.001
Park, 2020, Prediction of Alzheimer's disease based on deep neural network by integrating gene expression and DNA methylation dataset, Expert Syst. Appl., 140, 10.1016/j.eswa.2019.112873
Nishiwaki, 2020, Gene selection from microarray data for Alzheimer's disease using random forest, cognitive Analytics: concepts, methodologies, tools, and applications, IGI Global, 1391
Nishiwaki, 2016, Finding a Disease-Related Gene from Microarray Data Using Random Forest, 542
Zafeiris, 2018, An artificial neural network integrated pipeline for biomarker discovery using Alzheimer's disease as a case study, Comput. Struct. Biotechnol. J., 16, 77, 10.1016/j.csbj.2018.02.001
Balamurugan, 2017, Alzheimer's disease diagnosis by using dimensionality reduction based on knn classifier, Biomed. Pharmacol. J., 10, 1823, 10.13005/bpj/1299
Bhowmik, 2017, Comparative study on selection of genes causing Alzheimer's Dementia using meta-heuristic approaches, Int. J. Pure Appl. Math., 115, 45
Kong, 2009, Independent component analysis of Alzheimer's DNA microarray gene expression data, Mol. Neurodegener., 4, 1, 10.1186/1750-1326-4-5
Shigemizu, 2019, A comparison of machine learning classifiers for dementia with Lewy bodies using miRNA expression data, BMC Med. Genom., 12, 1, 10.1186/s12920-019-0607-3
Kumar, 2015, Feature selection and classification of microarray data using MapReduce based ANOVA and K-nearest neighbor, Procedia Comput. Sci., 54, 301, 10.1016/j.procs.2015.06.035
Shah, 2020, Optimized gene selection and classification of cancer from microarray gene expression data using deep learning, Neural Comput. Appl., 1
Urda, 2017, 50
Schmauch, 2020, A deep learning model to predict RNA-Seq expression of tumours from whole slide images, Nat. Commun., 11, 1, 10.1038/s41467-020-17678-4
Maj, 2019, Integration of machine learning methods to dissect genetically imputed transcriptomic profiles in Alzheimer's disease, Front. Genet., 10, 726, 10.3389/fgene.2019.00726
An, 2020
Beebe-Wang, 2018, MD-AD: multi-task deep learning for Alzheimer's disease neuropathology, bioRxiv
Wold, 1987, Principal component analysis, Chemometr. Intell. Lab. Syst., 2, 37, 10.1016/0169-7439(87)80084-9
Abdi, 2010, Principal component analysis, Wiley, Interdiscip. Rev. Comput. Stat., 2, 433, 10.1002/wics.101
González-Domínguez, 2012, Metabolomic approach to Alzheimer's disease diagnosis based on mass spectrometry, Chem. Pap., 66, 829, 10.2478/s11696-012-0184-9
Yuen, 2021, Prediction of differentially expressed microRNAs in blood as potential biomarkers for Alzheimer's disease by meta-analysis and adaptive boosting ensemble learning, Alzheimer's Res. Ther., 13, 1, 10.1186/s13195-021-00862-z
Cheng, 2021, Machine learning compensates fold-change method and highlights oxidative phosphorylation in the brain transcriptome of Alzheimer's disease, Sci. Rep., 11, 1
Zhao, 2019, Adaptive boosting-based computational model for predicting potential miRNA-disease associations, Bioinformatics, 35, 4730, 10.1093/bioinformatics/btz297
Madar, 2021, Identification of marker genes in Alzheimer's disease using a machine-learning model, Bioinformation, 17, 348, 10.6026/97320630017363
Cummings, 2018, Lessons learned from Alzheimer disease: clinical trials with negative outcomes, Clin. Transl. Sci., 11, 147, 10.1111/cts.12491
Ferreira, 2018, The heterogeneity within Alzheimer's disease, Aging, 10, 3058, 10.18632/aging.101638
Altman, 2018, The curse (s) of dimensionality, Nat. Methods, 15, 399, 10.1038/s41592-018-0019-x
Bzdok, 2018, Points of significance: statistics versus machine learning, Nat. Methods, 2018a, 1
Langfelder, 2017
Huang, 2009, Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists, Nucleic Acids Res., 37, 1, 10.1093/nar/gkn923
Duan, 2004, 49
Ma, 2015, Priori information based support vector regression and its applications, Math. Probl Eng., 10.1155/2015/974542
Das, 2020, sigFeature: novel significant feature selection method for classification of gene expression data using support vector machine and t statistic, Front. Genet., 11, 247, 10.3389/fgene.2020.00247
Zhang, 2003, Support vector machine classifications for microarray expression data set, 67
Díaz-Uriarte, 2006, Gene selection and classification of microarray data using random forest, BMC Bioinf., 7, 1, 10.1186/1471-2105-7-3
Monk, 2021, A machine learning method to identify genetic variants potentially associated with Alzheimer's disease, Front. Genet., 12, 10.3389/fgene.2021.647436
Jamil, 2020, Systematic multi-omics integration (MOI) approach in plant systems biology, Front. Plant Sci., 11, 944, 10.3389/fpls.2020.00944
Liu, 2010, Identifying dysfunctional crosstalk of pathways in various regions of Alzheimer's disease brains, BMC Syst. Biol., 1
Sood, 2015, A novel multi-tissue RNA diagnostic of healthy ageing relates to cognitive health status, Genome Biol., 16, 1, 10.1186/s13059-015-0750-x
Dey, 2019, Deep undepleted human serum proteome profiling toward biomarker discovery for Alzheimer's disease, Clin. Proteonomics, 16, 1
Ludwig, 2019, Machine learning to detect Alzheimer's disease from circulating non-coding RNAs, Dev. Reprod. Biol., 17, 430
Wang, 2019, Detecting diagnostic biomarkers of Alzheimer's disease by integrating gene expression data in six brain regions, Front. Genet., 10, 157, 10.3389/fgene.2019.00157
Zhao, 2020, A machine learning approach to identify a circulating MicroRNA signature for Alzheimer disease, J Appl. Lab. Med., 5, 15, 10.1373/jalm.2019.029595
Shigemizu, 2020, Identification of potential blood biomarkers for early diagnosis of Alzheimer's disease through RNA sequencing analysis, Alzheimer's Res. Ther., 12, 1, 10.1186/s13195-020-00654-x
Hasin, 2017, Multi-omics approaches to disease, Genome Biol., 18, 1, 10.1186/s13059-017-1215-1
Califano, 2012, Leveraging models of cell regulation and GWAS data in integrative network-based association studies, Nat. Genet., 44, 841, 10.1038/ng.2355
Argelaguet, 2018, Multi‐Omics factor analysis—a framework for unsupervised integration of multi‐omics data sets, Mol. Syst. Biol., 14, 10.15252/msb.20178124
Chaudhary, 2018, Deep learning–based multi-omics integration robustly predicts survival in liver cancer, Clin. Cancer Res., 24, 1248, 10.1158/1078-0432.CCR-17-0853
Satoh, 2017, 2513
Zhu, 2017, Integrating clinical and multiple omics data for prognostic assessment across human cancers, Sci. Rep., 7, 1, 10.1038/s41598-017-17031-8
Madhavan, 2013, Genome-wide multi-omics profiling of colorectal cancer identifies immune determinants strongly associated with relapse, Front. Genet., 4, 236, 10.3389/fgene.2013.00236
Kel, 2016, Multi-omics “upstream analysis” of regulatory genomic regions helps identifying targets against methotrexate resistance of colon cancer, EuPA Open Proteom, 13, 1, 10.1016/j.euprot.2016.09.002
Chen, 2017, APOBEC3A is an oral cancer prognostic biomarker in Taiwanese carriers of an APOBEC deletion polymorphism, Nat. Commun., 8, 1
Zhou, 2019, Longitudinal multi-omics of host–microbe dynamics in prediabetes, Nature, 569, 663, 10.1038/s41586-019-1236-x
Lloyd-Price, 2019, Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases, Nature, 569, 655, 10.1038/s41586-019-1237-9
Miao, 2014, Identification of prognostic biomarkers in hepatitis B virus-related hepatocellular carcinoma and stratification by integrative multi-omics analysis, J. Hepatol., 61, 840, 10.1016/j.jhep.2014.05.025
Cisek, 2016, The application of multi-omics and systems biology to identify therapeutic targets in chronic kidney disease, Nephrol. Dial. Transplant., 31, 2003, 10.1093/ndt/gfv364
Kaczorowski, 2019, Multi-omics analysis identifies gene networks associated with cognitive aging and Alzheimer's disease, Innov. Aging, 3, S587
Kohl, 2014, A practical data processing workflow for multi-OMICS projects, Biochim. Biophys. Acta Protein Proteonomics, 1844, 52, 10.1016/j.bbapap.2013.02.029
Shen, 2009, Integrative clustering of multiple genomic data types using a joint latent variable model with application to breast and lung cancer subtype analysis, Bioinformatics, 25, 2906, 10.1093/bioinformatics/btp543
Motsinger-Reif, 2013, Comparing metabolomic and pathologic biomarkers alone and in combination for discriminating Alzheimer's disease from normal cognitive aging, Acta Neuropathol. Commun., 1, 1, 10.1186/2051-5960-1-28
Rodrigues, 2017, Wrappers feature selection in Alzheimer's biomarkers using kNN and SMOTE oversampling, TeMA, 18, 15, 10.5540/tema.2017.018.01.0015
Park, 2015, Sparse overlapping group lasso for integrative multi-omics analysis, J. Comput. Biol., 22, 73, 10.1089/cmb.2014.0197
Clark, 2021, An integrative multi-omics approach reveals new central nervous system pathway alterations in Alzheimer's disease, Alzheimer's Res. Ther., 13, 1, 10.1186/s13195-021-00814-7
Hodson, 2016, Precision medicine, Nature, 537, 10.1038/537S49a
Forloni, 2020, Alzheimer's disease: from basic science to precision medicine approach, BMJ Neurol. Open, 2, 10.1136/bmjno-2020-000079
Berkowitz, 2018, Precision medicine for Alzheimer's disease prevention, Healthcare, Multidiscip. Digit. Publ. Inst., 82
Toups, 2021
Li, 2019, Epigenetic dysregulation of enhancers in neurons is associated with Alzheimer's disease pathology and cognitive symptoms, Nat. Commun., 10, 1
Jiang, 2017, The gut microbiota and Alzheimer's disease, J. Alzheim. Dis., 58, 1, 10.3233/JAD-161141
Jin, 2021, scGRNom: a computational pipeline of integrative multi-omics analyses for predicting cell-type disease genes and regulatory networks, Genome Med., 13, 1, 10.1186/s13073-021-00908-9
Gilligan, 2012, Racial and ethnic disparities in Alzheimer's disease pharmacotherapy exposure: an analysis across four state Medicaid populations, Am. J. Geriatr. Pharmacother., 10, 303, 10.1016/j.amjopharm.2012.09.002
Hallman, 1991, The apolipoprotein E polymorphism: a comparison of allele frequencies and effects in nine populations, Am. J. Hum. Genet., 49, 338
An, 2016, A genetic screen of the mutations in the Korean patients with early-onset Alzheimer's disease, Clin. Interv. Aging, 11, 1817, 10.2147/CIA.S116724
Zhou, 2018, Identification of genetic risk factors in the Chinese population implicates a role of immune system in Alzheimer's disease pathogenesis, Proc. Natl. Acad. Sci. Unit. States Am., 115, 1697, 10.1073/pnas.1715554115