Skip to main content

Systems Biology Methods for Alzheimer’s Disease Research Toward Molecular Signatures, Subtypes, and Stages and Precision Medicine: Application in Cohort Studies and Trials

  • Protocol
  • First Online:
Biomarkers for Alzheimer’s Disease Drug Development

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1750))

Abstract

Alzheimer’s disease (AD) is a complex multifactorial disease, involving a combination of genomic, interactome, and environmental factors, with essential participation of (a) intrinsic genomic susceptibility and (b) a constant dynamic interplay between impaired pathways and central homeostatic networks of nerve cells. The proper investigation of the complexity of AD requires new holistic systems-level approaches, at both the experimental and computational level. Systems biology methods offer the potential to unveil new fundamental insights, basic mechanisms, and networks and their interplay. These may lead to the characterization of mechanism-based molecular signatures, and AD hallmarks at the earliest molecular and cellular levels (and beyond), for characterization of AD subtypes and stages, toward targeted interventions according to the evolving precision medicine paradigm. In this work, an update on advanced systems biology methods and strategies for holistic studies of multifactorial diseases—particularly AD—is presented. This includes next-generation genomics, neuroimaging and multi-omics methods, experimental and computational approaches, relevant disease models, and latest genome editing and single-cell technologies. Their progressive incorporation into basic research, cohort studies, and trials is beginning to provide novel insights into AD essential mechanisms, molecular signatures, and markers toward mechanism-based classification and staging, and tailored interventions. Selected methods which can be applied in cohort studies and trials, with the European Prevention of Alzheimer’s Dementia (EPAD) project as a reference example, are presented and discussed.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 69.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 89.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 119.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Castrillo JI, Oliver SG (2011) Yeast systems biology. Methods and protocols. Methods in molecular biology 759 (MiMB series. Editor-in-chief. Prof. John M. Walker). Humana Press/Springer, New York

    Google Scholar 

  2. Castrillo JI, Oliver SG (2016a) Alzheimer’s as a systems-level disease involving the interplay of multiple cellular networks. Methods Mol Biol 1303:3–48

    Article  PubMed  Google Scholar 

  3. Castrillo JI, Oliver SG (2016b) Systems biology of Alzheimer’s disease. Methods in molecular biology (MiMB) series. Humana Press/Springer, New York

    Book  Google Scholar 

  4. Chen R, Mias GI, Li-Pook-Than J et al (2012) Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell 148:1293–1307

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Castrillo JI, Pir P, Oliver SG (2013) Yeast systems biology: towards a systems understanding of regulation of eukaryotic networks in complex diseases and biotechnology. In: Walhout M, Vidal M, Dekker J (eds) Handbook of systems biology. Elsevier, New York, pp 343–365

    Chapter  Google Scholar 

  6. Walhout M, Vidal M, Dekker J (2013) Handbook of systems biology. Elsevier, New York

    Google Scholar 

  7. Kosik KS (2015) Personalized medicine for effective Alzheimer disease treatment. JAMA Neurol 72:497–498

    Article  PubMed  Google Scholar 

  8. Montine TJ, Montine KS (2015) Precision medicine: clarity for the clinical and biological complexity of Alzheimer’s and Parkinson’s diseases. J Exp Med 212:601–605

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Kovacs GG (2016) Molecular pathological classification of neurodegenerative diseases: turning towards precision medicine. Int J Mol Sci 17:pii: E189

    Article  CAS  Google Scholar 

  10. Reitz C (2016) Toward precision medicine in Alzheimer’s disease. Ann Transl Med 4:107

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Swanton C, Soria JC, Bardelli A et al (2016) Consensus on precision medicine for metastatic cancers: a report from the MAP conference. Ann Oncol 27:1443–1448

    Article  PubMed  CAS  Google Scholar 

  12. Hampel H, O’Bryant SE, Castrillo JI et al (2016) Precision medicine - the golden gate to detect, prevent and cure Alzheimer’s disease. J Prev Alz Dis 3:243–259

    CAS  Google Scholar 

  13. Hampel H, O’Bryant SE, Durrleman S, Alzheimer Precision Medicine Initiative et al (2017) A precision medicine initiative for Alzheimer’s disease: the road ahead to biomarker-guided integrative disease modeling. Climacteric 20:107–118

    Article  PubMed  CAS  Google Scholar 

  14. Collins FS, Varmus H (2015) A new initiative on precision medicine. N Engl J Med 372:793–795

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Goodwin S, McPherson JD, McCombie WR (2016) Coming of age: ten years of next-generation sequencing technologies. Nat Rev Genet 17:333–351

    Article  PubMed  CAS  Google Scholar 

  16. Berg J (2016) Gene-environment interplay. Science 354:15

    Article  PubMed  CAS  Google Scholar 

  17. Kitano H (2002) Systems biology: a brief overview. Science 295:1662–1664

    Article  PubMed  CAS  Google Scholar 

  18. Castrillo JI, Oliver SG (2006) Metabolomics and systems biology in Saccharomyces cerevisiae. In: Karl Esser K (ed) The mycota. A comprehensive treatise on fungi as experimental systems for basic and applied research, Fungal genomics, vol XIII. Springer, New York, pp 3–18

    Google Scholar 

  19. Castrillo JI, Oliver SG (2014) Yeast as a model for systems Biology studies on complex Diseases. In: Nowrousian M (ed) The mycota. A comprehensive treatise on fungi as experimental systems for basic and applied research. Fungal genomics, 2nd edn (Karl Esser, Series Editor). Springer, Berlin, pp 3–30

    Google Scholar 

  20. Hampel H, Frank R, Broich K et al (2010) Biomarkers for Alzheimer’s disease: academic, industry and regulatory perspectives. Nat Rev Drug Discov 9:560–574

    Article  PubMed  CAS  Google Scholar 

  21. Hampel H, Lista S, Khachaturian ZS (2012) Development of biomarkers to chart all Alzheimer’s disease stages: the royal road to cutting the therapeutic Gordian Knot. Alzheimers Dement 8:312–336

    Article  PubMed  CAS  Google Scholar 

  22. Hampel H, Lista S (2013) Use of biomarkers and imaging to assess pathophysiology, mechanisms of action and target engagement. J Nutr Health Aging 17:54–63

    Article  PubMed  CAS  Google Scholar 

  23. Hampel H, Lista S, Teipel SJ et al (2014) Perspective on future role of biological markers in clinical therapy trials of Alzheimer’s disease: a long-range point of view beyond 2020. Biochem Pharmacol 88:426–449

    Article  PubMed  CAS  Google Scholar 

  24. Lista S, Emanuele E (2011) Role of amyloid β1-42 and neuroimaging biomarkers in Alzheimer’s disease. Biomark Med 5:411–413

    Article  PubMed  CAS  Google Scholar 

  25. Lista S, Garaci FG, Ewers M et al (2014) CSF Aβ1-42 combined with neuroimaging biomarkers in the early detection, diagnosis and prediction of Alzheimer’s disease. Alzheimers Dement 10:381–392

    Article  PubMed  Google Scholar 

  26. Zetzsche T, Rujescu D, Hardy J, Hampel H (2010) Advances and perspectives from genetic research: development of biological markers in Alzheimer’s disease. Expert Rev Mol Diagn 10:667–690

    Article  PubMed  CAS  Google Scholar 

  27. Bertram L, Hampel H (2011) The role of genetics for biomarker development in neurodegeneration. Prog Neurobiol 95:501–504

    Article  PubMed  CAS  Google Scholar 

  28. Hampel H, Lista S (2012) Alzheimer disease: from inherited to sporadic AD-crossing the biomarker bridge. Nat Rev Neurol 8:598–600

    Article  PubMed  Google Scholar 

  29. Lista S, O’Bryant SE, Blennow K et al (2015) Biomarkers in sporadic and familial Alzheimer’s disease. J Alzheimers Dis 47:291–317

    Article  PubMed  Google Scholar 

  30. Blennow K, Hampel H, Weiner M, Zetterberg H (2010) Cerebrospinal fluid and plasma biomarkers in Alzheimer disease. Nat Rev Neurol 6:131–144

    Article  PubMed  CAS  Google Scholar 

  31. Blennow K, Dubois B, Fagan AM et al (2015) Clinical utility of cerebrospinal fluid biomarkers in the diagnosis of early Alzheimer’s disease. Alzheimers Dement 11:58–69

    Article  PubMed  Google Scholar 

  32. Ghidoni R, Benussi L, Paterlini A et al (2011) Cerebrospinal fluid biomarkers for Alzheimer’s disease: the present and the future. Neurodegener Dis 8:413–420

    Article  PubMed  CAS  Google Scholar 

  33. Ewers M, Mattsson N, Minthon L et al (2015) CSF biomarkers for the differential diagnosis of Alzheimer’s disease: a large-scale international multicenter study. Alzheimers Dement 11:1306–1315

    Article  PubMed  Google Scholar 

  34. Lista S, Faltraco F, Prvulovic D, Hampel H (2013) Blood and plasma-based proteomic biomarker research in Alzheimer’s disease. Prog Neurobiol 101–102:1–17

    Article  PubMed  CAS  Google Scholar 

  35. Lista S, Faltraco F, Hampel H (2013) Biological and methodical challenges of blood-based proteomics in the field of neurological research. Prog Neurobiol 101–102:18–34

    Article  PubMed  CAS  Google Scholar 

  36. O’Bryant SE, Lista S, Rissman RA et al (2015) Comparing biological markers of Alzheimer’s disease across blood fraction and platforms: comparing apples to oranges. Alzheimers Dement (Amst) 3:27–34

    Google Scholar 

  37. O’Bryant SE, Gupta V, Henriksen K, STAR-B and BBBIG Working Groups et al (2015) Guidelines for the standardization of preanalytic variables for blood-based biomarker studies in Alzheimer’s disease research. Alzheimers Dement 11:549–560

    Article  PubMed  Google Scholar 

  38. O’Bryant SE, Mielke MM, Rissman RA et al (2017) Biofluid based biomarker professional interest area. Blood-based biomarkers in Alzheimer disease: current state of the science and a novel collaborative paradigm for advancing from discovery to clinic. Alzheimers Dement 13:45–58

    Article  PubMed  Google Scholar 

  39. Ewers M, Sperling RA, Klunk WE et al (2011) Neuroimaging markers for the prediction and early diagnosis of Alzheimer’s disease dementia. Trends Neurosci 34:430–442

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Teipel SJ, Grothe M, Lista S et al (2013) Relevance of magnetic resonance imaging for early detection and diagnosis of Alzheimer disease. Med Clin North Am 97:399–424

    Article  PubMed  Google Scholar 

  41. Villa A (2016) Book review (Systems biology of Alzheimer’s disease. Castrillo JI, Oliver SG (eds). Humana, Springer, New York, 2016). J Alzheimers Dis 50(4):1255–1256

    Article  Google Scholar 

  42. Braak H, Braak E, Bohl J, Bratzke H (1998) Evolution of Alzheimer’s disease related cortical lesions. J Neural Transm Suppl 54:97–106

    Article  PubMed  CAS  Google Scholar 

  43. Yankner BA, Lu T, Loerch P (2008) The aging brain. Annu Rev Pathol 3:41–66

    Article  PubMed  CAS  Google Scholar 

  44. Wood H (2014) Alzheimer disease: functional connectivity changes show similar trajectories in autosomal dominant and sporadic Alzheimer disease. Nat Rev Neurol 10:483

    Article  PubMed  Google Scholar 

  45. Blennow K, de Leon MJ, Zetterberg H (2006) Alzheimer’s disease. Lancet 368:387–403

    Article  PubMed  CAS  Google Scholar 

  46. Acosta-Baena N, Sepulveda-Falla D, Lopera-Gómez CM et al (2011) Pre-dementia clinical stages in presenilin 1 E280A familial early-onset Alzheimer’s disease: a retrospective cohort study. Lancet Neurol 10:213–220

    Article  PubMed  CAS  Google Scholar 

  47. Waring SC, Rosenberg RN (2008) Genome-wide association studies in Alzheimer disease. Arch Neurol 65:329–334

    Article  PubMed  Google Scholar 

  48. Alzheimer’s Society UK (2014) What is Alzheimer’s disease? Alzheimers.org.uk. https://www.alzheimers.org.uk/download/downloads/id/3379/what_is_alzheimers_disease.pdf

  49. Strittmatter WJ, Saunders AM, Schmechel D et al (1993) Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci U S A 90:1977–1981

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Guerreiro R, Brás J, Hardy J (2013) SnapShot: genetics of Alzheimer’s disease. Cell 155:968–968.e1

    Article  PubMed  CAS  Google Scholar 

  51. Budson AE, Kowall NW (2011) The handbook of Alzheimer’s disease and other dementias. Willey-Blackwell, New York

    Book  Google Scholar 

  52. Eisenstein M (2011) Genetics: finding risk factors. Nature 475:S20–S22. http://www.nature.com/nature/journal/v475/n7355_supp/full/475S20a.html

    Article  PubMed  CAS  Google Scholar 

  53. Benitez BA, Jin SC, Guerreiro R et al (2014) Missense variant in TREML2 protects against Alzheimer’s disease. Neurobiol Aging 35:1510.e19–1510.e26

    Article  CAS  Google Scholar 

  54. St George-Hyslop PH, Petit A (2005) Molecular biology and genetics of Alzheimer’s disease. C R Biol 328:119–130

    Article  PubMed  CAS  Google Scholar 

  55. Morgan K, Carrasquillo MM (2013) Genetic variants in Alzheimer’s disease. Springer, New York

    Book  Google Scholar 

  56. Karch CM, Cruchaga C, Goate AM (2014) Alzheimer’s disease genetics: from the bench to the clinic. Neuron 83:11–26

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Novarino G, Fenstermaker AG, Zaki MS et al (2014) Exome sequencing links corticospinal motor neuron disease to common neurodegenerative disorders. Science 343:506–511

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Tanzi RE (2014) Decoding Alzheimer’s in the age of genome-wide analyses. Mol Neurodeg 8(Suppl 1):O1

    Google Scholar 

  59. Bertram L (2016) Next generation sequencing in Alzheimer’s disease. Methods Mol Biol 1303:281–297

    Article  PubMed  Google Scholar 

  60. Jin SC, Benitez BA, Deming Y, Cruchaga C (2016) Pooled-DNA sequencing for elucidating new genomic risk factors, rare variants underlying Alzheimer’s disease. Methods Mol Biol 1303:299–314

    Article  PubMed  Google Scholar 

  61. Szigeti K (2016) New genome-wide methods for elucidation of candidate copy number variations (CNVs) contributing to Alzheimer’s disease heritability. Methods Mol Biol 1303:315–326

    Article  PubMed  Google Scholar 

  62. Rader DJ, Damrauer SM (2016) “Pheno”menal value for human health. Science 354:1534–1536

    Article  PubMed  CAS  Google Scholar 

  63. Huddleston J, Chaisson MJ, Meltz Steinberg K et al (2017) Discovery and genotyping of structural variation from long-read haploid genome sequence data. Genome Res 27:677–685

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Seo JS, Rhie A, Kim J et al (2016) De novo assembly and phasing of a Korean human genome. Nature 538:243–247

    Article  PubMed  CAS  Google Scholar 

  65. Chiang C, Scott AJ, Davis JR et al (2017) The impact of structural variation on human gene expression. Nat Genet 49(5):692–699. https://doi.org/10.1038/ng.3834

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Weisenfeld NI, Kumar V, Shah P et al (2017) Direct determination of diploid genome sequences. Genome Res 27(5):757–767. https://doi.org/10.1101/gr.214874.116

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Bickhart DM, Rosen BD, Koren S et al (2017) Single-molecule sequencing and chromatin conformation capture enable de novo reference assembly of the domestic goat genome. Nat Genet 49:643–650

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Worley KC (2017) A golden goat genome. Nat Genet 49:485–486

    Article  PubMed  CAS  Google Scholar 

  69. Dudchenko O, Batra SS, Omer AD et al (2017) De novo assembly of the Aedes aegypti genome using Hi-C yields chromosome-length scaffolds. Science 356(6333):92–95. https://doi.org/10.1126/science.aal3327

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Check Hayden E (2016) A radical revision of human genetics. Nature 53:154–157

    Google Scholar 

  71. Patra B, Kon Y, Yadav G et al (2017) A genome wide dosage suppressor network reveals genomic robustness. Nucleic Acids Res 45:255–270

    Article  PubMed  CAS  Google Scholar 

  72. Lek M, Karczewski KJ, Minikel EV, Exome Aggregation Consortium et al (2016) Analysis of protein-coding genetic variation in 60,706 humans. Nature 536:285–291

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Cummings BB, Marshall JL, Tukiainen T et al (2017) Improving genetic diagnosis in Mendelian disease with transcriptome sequencing. Sci Transl Med 9(386). https://doi.org/10.1126/scitranslmed.aal5209

    Article  PubMed  PubMed Central  Google Scholar 

  74. Baker-Nigh AT, Mawuenyega KG, Bollinger JG et al (2016) Human central nervous system (CNS) ApoE isoforms are increased by age, differentially altered by amyloidosis, and relative amounts reversed in the CNS compared with plasma. J Biol Chem 291:27204–27218

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Huang YA, Zhou B, Wernig M, Südhof TC (2017) ApoE2, ApoE3, and ApoE4 differentially stimulate APP transcription and Aβ secretion. Cell 168:427–441.e21

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Klann TS, Black JB, Chellappan M et al (2017) CRISPR-Cas9 epigenome editing enables high-throughput screening for functional regulatory elements in the human genome. Nat Biotechnol 35:561–568. https://doi.org/10.1038/nbt.3853

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. McConnell MJ, Moran JV, Abyzov A, The Brain Somatic Mosaicism Network et al (2017) Intersection of diverse neuronal genomes and neuropsychiatric disease: The Brain Somatic Mosaicism Network. Science 356(6336). https://doi.org/10.1126/science.aal1641

  78. Van der Flier WM (2016) Clinical heterogeneity in familial Alzheimer’s disease. Lancet Neurol 15:1296–1298

    Article  PubMed  Google Scholar 

  79. Hatami A, Monjazeb S, Milton S, Glabe CG (2017) Familial Alzheimer’s disease mutations within the amyloid precursor protein alter the aggregation and conformation of the amyloid-β peptide. J Biol Chem 292:3172–3185

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Senthil G, Dutka T, Bingaman L, Lehner T (2017) Genomic resources for the study of neuropsychiatric disorders. Mol Psychiatry 22:1659–1663. https://doi.org/10.1038/mp.2017.29

    Article  PubMed  CAS  Google Scholar 

  81. Litton JE (2017) We must urgently clarify data-sharing rules. Nature 541:437

    Article  PubMed  CAS  Google Scholar 

  82. Editorial (2016) The power of big data must be harnessed for medical progress. Nature 539:467–468

    Google Scholar 

  83. Auffray C, Balling R, Barroso I et al (2016) Making sense of big data in health research: towards an EU action plan. Genome Med 8:71

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Dubois B, Feldman HH, Jacova C et al (2014) Advancing research diagnostic criteria for Alzheimer’s disease: the IWG-2 criteria. Lancet Neurol 13:614–629

    Article  PubMed  Google Scholar 

  85. Mar J, Soto-Gordoa M, Arrospide A, Moreno-Izco F, Martínez-Lage P (2015) Fitting the epidemiology and neuropathology of the early stages of Alzheimer’s disease to prevent dementia. Alzheimers Res Ther 7(1):2. https://doi.org/10.1186/s13195-014-0079-9

    Article  PubMed  PubMed Central  Google Scholar 

  86. Jack CR Jr, Bennett DA, Blennow K et al (2016) A/T/N: an unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology 87:539–547

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Jack CR Jr, Wiste HJ, Weigand SD et al (2017) Age-specific and sex-specific prevalence of cerebral β-amyloidosis, tauopathy, and neurodegeneration in cognitively unimpaired individuals aged 50–95 years: a cross-sectional study. Lancet Neurol 16:435–444. https://doi.org/10.1016/S1474-4422(17)30077-7

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Johnson KA, Minoshima S, Bohnen NI et al (2013) Appropriate use criteria for amyloid PET: a report of the amyloid imaging task force, the Society of Nuclear Medicine and Molecular Imaging, and the Alzheimer’s Association. Alzheimers Dement 9:e-1–e16

    Article  Google Scholar 

  89. Mak E, Gabel S, Mirette H et al (2016) Structural neuroimaging in preclinical dementia: from microstructural deficits and grey matter atrophy to macroscale connectomic changes. Ageing Res Rev 35:250–264. https://doi.org/10.1016/j.arr.2016.10.001

    Article  PubMed  Google Scholar 

  90. Nichols TE, Das S, Eickhoff SB et al (2017) Best practices in data analysis and sharing in neuroimaging using MRI. Nat Neurosci 20:299–303

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Sepulcre J, Masdeu JC (2016) Advanced neuroimaging methods towards characterization of early stages of Alzheimer’s disease. Methods Mol Biol 1303:509–519

    Article  PubMed  Google Scholar 

  92. Masdeu JC (2017) Future directions in imaging neurodegeneration. Curr Neurol Neurosci Rep 17:9

    Article  PubMed  CAS  Google Scholar 

  93. Sepulcre J, Grothe MJ, Sabuncu M et al (2017) Hierarchical organization of tau and amyloid deposits in the cerebral cortex. JAMA Neurol 74:813–820. https://doi.org/10.1001/jamaneurol.2017.0263

    Article  PubMed  PubMed Central  Google Scholar 

  94. Vemuri P, Schöll M (2017) Linking amyloid-β and tau deposition in Alzheimer disease. JAMA Neurol 74:766–768. https://doi.org/10.1001/jamaneurol.2017.0323

    Article  PubMed  Google Scholar 

  95. Matsuda H, Asada T, Tokumaru AM (2017) Neuroimaging diagnosis for Alzheimer’s disease and other dementias. Springer, Tokyo

    Book  Google Scholar 

  96. Fan Z, Brooks DJ, Okello A, Edison P (2017) An early and late peak in microglial activation in Alzheimer’s disease trajectory. Brain 140:792–803. https://doi.org/10.1093/brain/aww349

    Article  PubMed  PubMed Central  Google Scholar 

  97. Pickett EK, Henstridge CM, Allison E et al (2017) Spread of tau down neural circuits precedes synapse and neuronal loss in the rTgTauEC mouse model of early Alzheimer’s disease. Synapse. https://doi.org/10.1002/syn.21965

  98. Brosch JR, Farlow MR, Risacher SL, Apostolova LG (2017) Tau imaging in Alzheimer’s disease diagnosis and clinical trials. Neurotherapeutics 14:62–68

    Article  PubMed  CAS  Google Scholar 

  99. Wang L, Benzinger TL, Su Y et al (2016) Evaluation of tau imaging in staging Alzheimer disease and revealing interactions between β-amyloid and tauopathy. JAMA Neurol 73:1070–1077

    Article  PubMed  PubMed Central  Google Scholar 

  100. Park JY, Na HK, Kim S, The Alzheimer’s Disease Neuroimaging Initiative et al (2017) Robust identification of Alzheimer’s disease subtypes based on cortical atrophy patterns. Sci Rep 7:43270

    Article  PubMed  PubMed Central  Google Scholar 

  101. Ossenkoppele R, Schonhaut DR, Schöll M et al (2016) Tau PET patterns mirror clinical and neuroanatomical variability in Alzheimer’s disease. Brain 139:1551–1567

    Article  PubMed  PubMed Central  Google Scholar 

  102. Xia C, Makaretz SJ, Caso C et al (2017) Association of in vivo [18F]AV-1451 tau PET imaging results with cortical atrophy and symptoms in typical and atypical Alzheimer disease. JAMA Neurol 74:427–436. https://doi.org/10.1001/jamaneurol.2016.5755

    Article  PubMed  PubMed Central  Google Scholar 

  103. Masdeu JC (2017b) Tau and cortical thickness in Alzheimer disease. JAMA Neurol 74:390–392. https://doi.org/10.1001/jamaneurol.2016.5701

    Article  PubMed  Google Scholar 

  104. Qian J, Hyman BT, Betensky RA (2017) Neurofibrillary tangle stage and the rate of progression of Alzheimer symptoms: Modeling using an autopsy cohort and application to clinical trial design. JAMA Neurol 74:540–548. https://doi.org/10.1001/jamaneurol.2016.5953

    Article  PubMed  PubMed Central  Google Scholar 

  105. Perneczky R, Tene O, Attems J et al (2016) Is the time ripe for new diagnostic criteria of cognitive impairment due to cerebrovascular disease? Consensus report of the international congress on vascular dementia working group. BMC Med 14:162

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Tan J, Li QX, Evin G (2016) Effects of mild and severe oxidative stress on BACE1 expression and APP amyloidogenic processing. Methods Mol Biol 1303:101–116

    Article  PubMed  Google Scholar 

  107. García-Osta A, Cuadrado-Tejedor M (2016) Advanced assay monitoring APP-carboxyl-terminal fragments as markers of APP processing in Alzheimer disease mouse models. Methods Mol Biol 1303:117–123

    Article  PubMed  Google Scholar 

  108. Pinotsi D, Kaminski Schierle GS, Kaminski CF (2016) Optical super-resolution imaging of β-amyloid aggregation in vitro and in vivo: method and techniques. Methods Mol Biol 1303:125–141

    Article  PubMed  Google Scholar 

  109. Rábano A, Cuadros R, Merino-Serráis P et al (2016) Protocols for monitoring the development of tau pathology in Alzheimer’s disease. Methods Mol Biol 1303:143–160

    Article  PubMed  Google Scholar 

  110. Kaufman SK, Sanders DW, Thomas TL et al (2016) Tau prion strains dictate patterns of cell pathology, progression rate, and regional vulnerability in vivo. Neuron 92:796–812

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  111. Hetz C, Mollereau B (2014) Disturbance of endoplasmic reticulum proteostasis in neurodegenerative diseases. Nat Rev Neurosci 15:233–249

    Article  PubMed  CAS  Google Scholar 

  112. Streeter A, Menzies FM, Rubinsztein DC (2016) LC3-II tagging and western blotting for monitoring autophagic activity in mammalian cells. Methods Mol Biol 1303:161–170

    Article  PubMed  Google Scholar 

  113. Feng T, Tammineni P, Agrawal C et al (2017) Autophagy-mediated regulation of BACE1 protein trafficking and degradation. J Biol Chem 292:1679–1690

    Article  PubMed  CAS  Google Scholar 

  114. Grimm A, Schmitt K, Eckert A (2016) Advanced mitochondrial respiration assay for evaluation of mitochondrial dysfunction in Alzheimer’s disease. Methods Mol Biol 1303:171–183

    Article  PubMed  Google Scholar 

  115. Gomez-Nicola D, Perry VH (2016) Analysis of microglial proliferation in Alzheimer’s disease. Methods Mol Biol 1303:185–193

    Article  PubMed  Google Scholar 

  116. Masters SL, O’Neill LA (2011) Disease-associated amyloid and misfolded protein aggregates activate the inflammasome. Trends Mol Med 17:276–282

    Article  PubMed  CAS  Google Scholar 

  117. McManus RM, Heneka MT (2017) Role of neuroinflammation in neurodegeneration: new insights. Alzheimers Res Ther 9:14

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  118. Jessen NA, Munk AS, Lundgaard I, Nedergaard M (2015) The glymphatic system: a beginner’s guide. Neurochem Res 40:2583–2599

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Zeppenfeld DM, Simon M, Haswell JD et al (2017) Association of perivascular localization of aquaporin-4 with cognition and Alzheimer disease in aging brains. JAMA Neurol 74:91–99

    Article  PubMed  Google Scholar 

  120. Tosto G, Reitz C (2016) Use of “omics” technologies to dissect neurologic disease. Handb Clin Neurol 138:91–106

    Article  PubMed  CAS  Google Scholar 

  121. Chen BJ, Mills JD, Janitz C, Janitz M (2016) RNA-sequencing to elucidate early patterns of dysregulation underlying the onset of Alzheimer’s disease. Methods Mol Biol 1303:327–347

    Article  PubMed  Google Scholar 

  122. Roth W, Hecker D, Fava E (2016) Systems biology approaches to the study of biological networks underlying Alzheimer’s disease: role of miRNAs. Methods Mol Biol 1303:349–377

    Article  PubMed  Google Scholar 

  123. Pichler S, Gu W, Hartl D et al (2017) The miRNome of Alzheimer’s disease: consistent downregulation of the miR-132/212 cluster. Neurobiol Aging 50:167.e1–167.e10

    Article  CAS  Google Scholar 

  124. Bai B, Hales CM, Chen PC et al (2013) U1 small nuclear ribonucleoprotein complex and RNA splicing alterations in Alzheimer’s disease. Proc Natl Acad Sci U S A 110:16562–16567

    Article  PubMed  PubMed Central  Google Scholar 

  125. Yin X, Jin N, Shi J et al (2017) Dyrk1A overexpression leads to increase of 3R-tau expression and cognitive deficits in Ts65Dn Down syndrome mice. Sci Rep 7(1):619. https://doi.org/10.1038/s41598-017-00682-y

    Article  PubMed  PubMed Central  Google Scholar 

  126. Hare DJ, Rembach A, Roberts BR (2016) The emerging role of metalloproteomics in Alzheimer’s disease research. Methods Mol Biol 1303:379–389

    Article  PubMed  Google Scholar 

  127. Di Domenico F, Perluigi M, Butterfield DA (2016) Redox proteomics in human biofluids: sample preparation, separation and immunochemical tagging for analysis of protein oxidation. Methods Mol Biol 1303:391–403

    Article  PubMed  Google Scholar 

  128. Nuzzo D, Inguglia L, Walters J et al (2017) A shotgun proteomics approach reveals a new toxic role for Alzheimer’s disease Aβ peptide: Spliceosome impairment. J Proteome Res 16:1526–1541. https://doi.org/10.1021/acs.jproteome.6b00925

    Article  PubMed  CAS  Google Scholar 

  129. Reinders J (2016) Proteomics in systems biology. Methods and protocols, Methods in molecular biology (MIMB) series. Humana Press/Springer, New York

    Book  Google Scholar 

  130. Perneczky R, Guo LH (2016) Plasma proteomics biomarkers in Alzheimer’s disease: latest advances and challenges. Methods Mol Biol 1303:521–529

    Article  PubMed  Google Scholar 

  131. Bai B, Chen PC, Hales CM et al (2014) Integrated approaches for analyzing U1-70K cleavage in Alzheimer’s disease. J Proteome Res 13:4526–4534

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Hales CM, Seyfried NT, Dammer EB et al (2014a) U1 small nuclear ribonucleoproteins (snRNPs) aggregate in Alzheimer’s disease due to autosomal dominant genetic mutations and trisomy 21. Mol Neurodegener 9:15

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  133. Hales CM, Dammer EB, Diner I et al (2014b) Aggregates of small nuclear ribonucleic acids (snRNAs) in Alzheimer’s disease. Brain Pathol 24:344–351

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  134. Kaddurah-Daouk R, Krishnan KR (2009) Metabolomics: a global biochemical approach to the study of central nervous system diseases. Neuropsychopharmacology 34:173–186

    Article  PubMed  CAS  Google Scholar 

  135. Kaddurah-Daouk R, Zhu H, Sharma S, Pharmacometabolomics Research Network et al (2013) Alterations in metabolic pathways and networks in Alzheimer’s disease. Transl Psychiatry 3:e244

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  136. Toledo JB, Arnold M, Kastenmüller G, The Alzheimer’s Disease Neuroimaging Initiative and the Alzheimer Disease Metabolomics Consortium et al (2017) Metabolic network failures in Alzheimer’s disease-A biochemical road map. Alzheimers Dement 13:965–984. https://doi.org/10.1016/j.jalz.2017.01.020

    Article  PubMed  PubMed Central  Google Scholar 

  137. Wang M, Han X (2016) Advanced shotgun lipidomics for characterization of altered lipid patterns in neurodegenerative diseases and brain injury. Methods Mol Biol 1303:405–422

    Article  PubMed  PubMed Central  Google Scholar 

  138. Proitsi P, Kim M, Whiley L et al (2017) Association of blood lipids with Alzheimer’s disease: a comprehensive lipidomics analysis. Alzheimers Dement 13:140–151

    Article  PubMed  Google Scholar 

  139. Huttlin EL, Bruckner RJ, Paulo JA et al (2017) Architecture of the human interactome defines protein communities and disease networks. Nature 545:505–509. https://doi.org/10.1038/nature22366

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Bennett RE, DeVos SL, Dujardin S et al (2017) Enhanced tau aggregation in the presence of amyloid β. Am J Pathol 187:1601–1612. https://doi.org/10.1016/j.ajpath.2017.03.011

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Kikuchi M, Ogishima S, Mizuno S et al (2016) Network-based analysis for uncovering mechanisms underlying Alzheimer’s disease. Methods Mol Biol 1303:479–491

    Article  PubMed  Google Scholar 

  142. Yerbury J, Bean D, Favrin G (2016) Network approaches to the understanding of Alzheimer’s disease: from model organisms to humans. Methods Mol Biol 1303:447–458

    Article  PubMed  Google Scholar 

  143. Zanzoni A (2016) A computational network biology approach to uncover novel genes related to Alzheimer’s disease. Methods Mol Biol 1303:435–446

    Article  PubMed  Google Scholar 

  144. Zhang B, Tran L, Emilsson V, Zhu J (2016) Characterization of genetic networks associated with Alzheimer’s disease. Methods Mol Biol 1303:459–477

    Article  PubMed  Google Scholar 

  145. Christianson JC, Olzmann JA, Shaler TA et al (2011) Defining human ERAD networks through an integrative mapping strategy. Nat Cell Biol 14:93–105

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. Kikuchi M, Ogishima S, Miyamoto T et al (2013) Identification of unstable network modules reveals disease modules associated with the progression of Alzheimer’s disease. PLoS One 8:e76162

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  147. Gitter A, Bar-Joseph Z (2016) The SDREM method for reconstructing signaling and regulatory response networks: applications for studying disease progression. Methods Mol Biol 1303:493–506

    Article  PubMed  Google Scholar 

  148. Genolini C, Ecochard R, Benghezal M et al (2016) kmlShape: an efficient method to cluster longitudinal data (time-series) according to their shapes. PLoS One 11:e0150738

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  149. Verduyckt M, Vignaud H, Bynens T et al (2016) Yeast as a model for Alzheimer’s disease: latest studies and advanced strategies. Methods Mol Biol 1303:197–215

    Article  PubMed  Google Scholar 

  150. Porzoor A, Macreadie I (2016) Yeast as a model for studies on Aβ aggregation toxicity in Alzheimer’s disease, autophagic responses, and drug screening. Methods Mol Biol 1303:217–226

    Article  PubMed  Google Scholar 

  151. Lim JY, Ott S, Crowther DC (2016) Drosophila melanogaster as a model for studies on the early stages of Alzheimer’s disease. Methods Mol Biol 1303:227–239

    Article  PubMed  Google Scholar 

  152. Cuadrado-Tejedor M, García-Osta A (2016) Chronic mild stress assay leading to early onset and propagation of Alzheimer’s disease phenotype in mouse models. Methods Mol Biol 1303:241–246

    Article  PubMed  Google Scholar 

  153. Lu M, Lawrence DA, Marsters S (2014b) Cell death. Opposing unfolded-protein-response signals converge on death receptor 5 to control apoptosis. Science 345:98–101

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  154. Abisambra JF, Jinwal UK, Blair LJ et al (2013) Tau accumulation activates the unfolded protein response by impairing endoplasmic reticulum-associated degradation. J Neurosci 33:9498–9507

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  155. Zhou L, McInnes J, Wierda K et al (2017) Tau association with synaptic vesicles causes presynaptic dysfunction. Nat Commun 8:15295. https://doi.org/10.1038/ncomms15295

    Article  PubMed  PubMed Central  Google Scholar 

  156. Kazim SF, Blanchard J, Bianchi R, Iqbal K (2017) Early neurotrophic pharmacotherapy rescues developmental delay and Alzheimer’s-like memory deficits in the Ts65Dn mouse model of Down syndrome. Sci Rep 7:45561. https://doi.org/10.1038/srep45561

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  157. Weick JP, Kang H, Bonadurer GF 3rd, Bhattacharyya A (2016) Gene expression studies on human Trisomy 21 iPSCs and neurons: towards mechanisms underlying Down’s syndrome and early Alzheimer’s disease-like pathologies. Methods Mol Biol 1303:247–265

    Article  PubMed  Google Scholar 

  158. Saurat NG, Livesey FJ, Moore S (2016) Cortical differentiation of human pluripotent cells for in vitro modeling of Alzheimer’s disease. Methods Mol Biol 1303:267–278

    Article  PubMed  Google Scholar 

  159. Choi SH, Kim YH, Hebisch M et al (2014) A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature 515:274–278

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  160. Choi SH, Kim YH, Quinti L et al (2016) 3D culture models of Alzheimer’s disease: a road map to a “cure-in-a-dish”. Mol Neurodegener 11:75

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  161. Camp JG, Treutlein B (2017) Human development: advances in mini-brain technology. Nature 545:39–40. https://doi.org/10.1038/545039a

    Article  PubMed  CAS  Google Scholar 

  162. Koo T, Lee J, Kim JS (2015) Measuring and reducing off-target activities of programmable nucleases including CRISPR-Cas9. Mol Cells 38:475–481

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  163. Schmid-Burgk JL, Chauhan D, Schmidt T et al (2016) A genome-wide CRISPR (clustered regularly interspaced short palindromic repeats) screen identifies NEK7 as an essential component of NLRP3 inflammasome activation. J Biol Chem 291:103–109

    Article  PubMed  CAS  Google Scholar 

  164. Paquet D, Kwart D, Chen A et al (2016) Efficient introduction of specific homozygous and heterozygous mutations using CRISPR/Cas9. Nature 533:125–129

    Article  PubMed  CAS  Google Scholar 

  165. Mungenast AE, Siegert S, Tsai LH (2016) Modeling Alzheimer’s disease with human induced pluripotent stem (iPS) cells. Mol Cell Neurosci 73:13–31

    Article  PubMed  CAS  Google Scholar 

  166. Owens B (2012) Genomics: the single life. Nature 491:27–29

    Article  PubMed  CAS  Google Scholar 

  167. Single-cell technology Focus Issue (2016) In this issue. Nat Biotechnol 34:vii. doi: https://doi.org/10.1038/nbt.3732

  168. De Strooper B, Karran E (2016) The cellular phase of Alzheimer’s disease. Cell 164:603–615

    Article  PubMed  CAS  Google Scholar 

  169. Ledford H (2017) The race to map the human body - one cell at a time. Nature 542:404–405

    Article  PubMed  CAS  Google Scholar 

  170. Xu Y, Mizuno T, Sridharan A et al (2016) Single-cell RNA sequencing identifies diverse roles of epithelial cells in idiopathic pulmonary fibrosis. JCI Insight 1:e90558

    Article  PubMed  PubMed Central  Google Scholar 

  171. Frazer S, Prados J, Niquille M et al (2017) Transcriptomic and anatomic parcellation of 5-HT(3A)R expressing cortical interneuron subtypes revealed by single-cell RNA sequencing. Nat Commun 8:14219. https://doi.org/10.1038/ncomms14219

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  172. Pavličev M, Wagner GP, Chavan AR et al (2017) Single-cell transcriptomics of the human placenta: inferring the cell communication network of the maternal-fetal interface. Genome Res 27:349–361

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  173. Zenobi R (2013) Single-cell metabolomics: analytical and biological perspectives. Science 342:1243259

    Article  PubMed  CAS  Google Scholar 

  174. Clyde D (2017) Technique: barcoding the nucleus. Nat Rev Genet 18:4–211. https://doi.org/10.1038/nrg.2017.11

    Article  CAS  Google Scholar 

  175. Ofengeim D, Giagtzoglou N, Huh D et al (2017) Single-cell RNA sequencing: unraveling the brain one cell at a time. Trends Mol Med 23:563–576. https://doi.org/10.1016/j.molmed.2017.04.006

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  176. Liang Y, Li K, Riecken K et al (2016) Long-term in vivo single-cell tracking reveals the switch of migration patterns in adult-born juxtaglomerular cells of the mouse olfactory bulb. Cell Res 26:805–821

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  177. Li H, Courtois ET, Sengupta D et al (2017) Reference component analysis of single-cell transcriptomes elucidates cellular heterogeneity in human colorectal tumors. Nat Genet 49:708–718. https://doi.org/10.1038/ng.3818

    Article  PubMed  CAS  Google Scholar 

  178. Keren-Shaul H, Spinrad A, Weiner A et al (2017) A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169:1276–1290.e17. https://doi.org/10.1016/j.cell.2017.05.018

    Article  PubMed  CAS  Google Scholar 

  179. Dubois B, Hampel H, Feldman HH et al (2016) Preclinical Alzheimer’s disease: definition, natural history, and diagnostic criteria. Alzheimers Dement 12:292–323

    Article  PubMed  PubMed Central  Google Scholar 

  180. Uzilov AV, Ding W, Fink MY et al (2016) Development and clinical application of an integrative genomic approach to personalized cancer therapy. Genome Med 8:62

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  181. Rajamani D, Bhasin MK (2016) Identification of key regulators of pancreatic cancer progression through multidimensional systems-level analysis. Genome Med 8:38

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  182. Ghosh D, Funk CC, Caballero J et al (2017) A cell-surface membrane protein signature for glioblastoma. Cell Syst 4:516–529

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  183. Sheridan C (2015) Omics-driven startups challenge healthcare model. Nat Biotechnol 33:887–889

    Article  PubMed  CAS  Google Scholar 

  184. Lausted C, Lee I, Zhou Y et al (2014) Systems approach to neurodegenerative disease biomarker discovery. Annu Rev Pharmacol Toxicol 54:457–481

    Article  PubMed  CAS  Google Scholar 

  185. Lista S, Khachaturian ZS, Rujescu D et al (2016) Application of systems theory in longitudinal studies on the origin and progression of Alzheimer’s disease. Methods Mol Biol 1303:49–67

    Article  PubMed  Google Scholar 

  186. Rollo JL, Banihashemi N, Vafaee F et al (2016) Unraveling the mechanistic complexity of Alzheimer’s disease through systems biology. Alzheimers Dement 12:708–718

    Article  PubMed  Google Scholar 

  187. Burton A (2016) Kaj Blennow: the route to biomarkers and the Söderberg prize. Lancet Neurol 15(9):906. https://doi.org/10.1016/S1474-4422(16)30097-7

    Article  PubMed  Google Scholar 

  188. Zwan MD, Rinne JO, Hasselbalch SG et al (2016) Use of amyloid-PET to determine cutpoints for CSF markers: a multicenter study. Neurology 86:50–58

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  189. FitzGerald GA (2016) Measure for measure: biomarker standards and transparency. Sci Transl Med 8:343fs10

    Article  PubMed  CAS  Google Scholar 

  190. Kivipelto M, Håkansson K (2017) A rare success against Alzheimer’s. Sci Am 316:32–37

    Article  PubMed  Google Scholar 

  191. Ritchie CW, Molinuevo JL, Satlin A et al (2016) The European Prevention of Alzheimer’s Dementia (EPAD) Consortium: a platform to enable the secondary prevention of Alzheimer’s dementia through improved Proof of Concept Trials. Lancet Psychiatry 3:179–186

    Article  PubMed  Google Scholar 

  192. Ritchie K, Ritchie CW, Yaffe K et al (2015) Is late-onset Alzheimer’s disease really a disease of midlife? Alzheimers Dement 1(2):122–130

    Google Scholar 

  193. Ritchie CW, Ames D, Clayton T, Lai R (2004) A meta-analysis of randomised trials for the efficacy and safety of donepezil, galantamine and rivastigmine for the treatment of Alzheimer’s disease. Am J Geriatr Psychiatry 12:358–369

    Article  PubMed  Google Scholar 

  194. Molinuevo JL, Jordi C, Came X et al (2016) Ethical challenges in preclinical Alzheimer’s disease observational studies and trials: results of the Barcelona Summit. Alzheimers Dement 12:614–622

    Article  PubMed  PubMed Central  Google Scholar 

  195. Ritchie K, Ropacki M, Albala B et al (2017) Recommended cognitive outcomes in preclinical Alzheimer’s disease: consensus statement from the European Prevention of Alzheimer’s Dementia project. Alzheimers Dement 13:186–195

    Article  PubMed  Google Scholar 

  196. Mortamais M, Ash JA, Harrison J et al (2017) Detecting cognitive changes in preclinical Alzheimer’s disease: a review of its feasibility. Alzheimers Dement 13:468–492

    Article  PubMed  Google Scholar 

  197. Bateman RJ, Xiong C, Benzinger TL, Dominantly Inherited Alzheimer Network et al (2012) Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. N Engl J Med 367:795–804

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  198. Fagan AM, Xiong C, Jasielec MS, Dominantly Inherited Alzheimer Network et al (2014) Longitudinal change in CSF biomarkers in autosomal-dominant Alzheimer’s disease. Sci Transl Med 6:226ra30

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  199. Imamura K, Izumi Y, Watanabe A et al (2017) The Src/c-Abl pathway is a potential therapeutic target in amyotrophic lateral sclerosis. Sci Transl Med 9. https://doi.org/10.1126/scitranslmed.aaf3962

  200. Sun C, Fang Y, Yin J et al (2017) Rational combination therapy with PARP and MEK inhibitors capitalizes on therapeutic liabilities in RAS mutant cancers. Sci Transl Med 9. https://doi.org/10.1126/scitranslmed.aal5148

  201. Cancer Genome Atlas Research Network (2017) Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell 169:1327–1341.e23

    Article  CAS  Google Scholar 

  202. Norton S, Matthews FE, Barnes DE et al (2014) Potential for primary prevention of Alzheimer’s disease: an analysis of population-based data. Lancet Neurol 13:788–794

    Article  PubMed  Google Scholar 

  203. World Alzheimer Report (2014) Dementia and risk reduction: an analysis of protective and modifiable factors. ADI (http://www.alz.co.uk/research/world-report-2014)

  204. Sperling RA, Karlawish J, Johnson KA (2013) Preclinical Alzheimer disease-the challenges ahead. Nat Rev Neurol 9:54–58

    Article  PubMed  CAS  Google Scholar 

  205. Epelbaum S, Genthon R, Cavedo E et al (2017) Preclinical Alzheimer’s disease: a systematic review of the cohorts underlying the concept. Alzheimers Dement 13:454–467

    Article  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported by Genetadi Biotech SL (Bizkaia, Spain). J.I.C. is the beneficiary of a senior prossgram (mode A) of Bizkaia:Xede Foundation. H.H. is supported by the AXA Research Fund, the Fondation Université Pierre et Marie Curie, and the Fondation pour la Recherche sur Alzheimer, Paris, France. Ce travail a bénéficié d’une aide de l’Etat “Investissements d’avenir” ANR-10-IAIHU-06 (H.H.). The research leading to these results has received funding from the program “Investissements d’avenir” ANR-10-IAIHU-06 (Agence Nationale de la Recherche-10-IA Agence Institut Hospitalo-Universitaire-6) (H.H.).

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer Science+Business Media, LLC

About this protocol

Check for updates. Verify currency and authenticity via CrossMark

Cite this protocol

Castrillo, J.I., Lista, S., Hampel, H., Ritchie, C.W. (2018). Systems Biology Methods for Alzheimer’s Disease Research Toward Molecular Signatures, Subtypes, and Stages and Precision Medicine: Application in Cohort Studies and Trials. In: Perneczky, R. (eds) Biomarkers for Alzheimer’s Disease Drug Development. Methods in Molecular Biology, vol 1750. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-7704-8_3

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-7704-8_3

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-7703-1

  • Online ISBN: 978-1-4939-7704-8

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics