Skip to main content

Application of Systems Theory in Longitudinal Studies on the Origin and Progression of Alzheimer’s Disease

  • Protocol
Systems Biology of Alzheimer's Disease

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1303))

Abstract

This chapter questions the prevailing “implicit” assumption that molecular mechanisms and the biological phenotype of dominantly inherited early-onset alzheimer’s disease (EOAD) could serve as a linear model to study the pathogenesis of sporadic late-onset alzheimer’s disease (LOAD). Now there is growing evidence to suggest that such reductionism may not be warranted; these suppositions are not adequate to explain the molecular complexities of LOAD. For example, the failure of some recent amyloid-centric clinical trials, which were largely based on the extrapolations from EOAD biological phenotypes to the molecular mechanisms in the pathogenesis of LOAD, might be due to such false assumptions. The distinct difference in the biology of LOAD and EOAD is underscored by the presence of EOAD cases without evidence of familial clustering or Mendelian transmission and, conversely, the discovery and frequent reports of such clustering and transmission patterns in LOAD cases. The primary thesis of this chapter is that a radically different way of thinking is required for comprehensive explanations regarding the distinct complexities in the molecular pathogenesis of inherited and sporadic forms of Alzheimer’s disease (AD). We propose using longitudinal analytical methods and the paradigm of systems biology (using transcriptomics, proteomics, metabolomics, and lipidomics) to provide us a more comprehensive insight into the lifelong origin and progression of different molecular mechanisms and neurodegeneration. Such studies should aim to clarify the role of specific pathophysiological and signaling pathways such as neuroinflammation, altered lipid metabolism, apoptosis, oxidative stress, tau hyperphosphorylation, protein misfolding, tangle formation, and amyloidogenic cascade leading to overproduction and reduced clearance of aggregating amyloid-beta (Aβ) species. A more complete understanding of the distinct difference in molecular mechanisms, signaling pathways, as well as comparability of the various forms of AD is of paramount importance. The development of knowledge and technologies for early detection and characterization of the disease across all stages will improve the predictions regarding the course of the disease, prognosis, and response to treatment. No doubt such advances will have a significant impact on the clinical management of both EOAD and LOAD patients. The approach propped here, combining longitudinal studies with the systems biology paradigm, will create a more effective and comprehensive framework for development of prevention therapies in AD.

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 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 249.99
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. Blennow K, De Leon MJ, Zetterberg H (2006) Alzheimer’s disease. Lancet 368:387–403

    CAS  PubMed  Google Scholar 

  2. Bertram L, Lill CM, Tanzi RE (2010) The genetics of Alzheimer disease: back to the future. Neuron 68:270–281

    CAS  PubMed  Google Scholar 

  3. Bertram L, Tanzi RE (2012) The genetics of Alzheimer’s disease. Prog Mol Biol Transl Sci 107:79–100

    CAS  PubMed  Google Scholar 

  4. Van Deerlin VM, Wood EM, Moore P et al (2007) Clinical, genetic, and pathologic characteristics of patients with frontotemporal dementia and progranulin mutations. Arch Neurol 64:1148–1153

    PubMed  Google Scholar 

  5. Huey ED, Grafman J, Wassermann EM et al (2006) Characteristics of frontotemporal dementia patients with a Progranulin mutation. Ann Neurol 60:374–380

    PubMed Central  PubMed  Google Scholar 

  6. Perry DC, Lehmann M, Yokoyama JS et al (2013) Progranulin mutations as risk factors for Alzheimer disease. JAMA Neurol 70:774–778

    PubMed Central  PubMed  Google Scholar 

  7. Jin SC, Pastor P, Cooper B et al (2012) Pooled-DNA sequencing identifies novel causative variants in PSEN1, GRN and MAPT in a clinical early-onset and familial Alzheimer’s disease Ibero-American cohort. Alzheimers Res Ther 4:34

    CAS  PubMed Central  PubMed  Google Scholar 

  8. Kelley BJ, Haidar W, Boeve BF et al (2010) Alzheimer disease-like phenotype associated with the c.154delA mutation in progranulin. Arch Neurol 67:171–177

    PubMed Central  PubMed  Google Scholar 

  9. Finch N, Baker M, Crook R et al (2009) Plasma progranulin levels predict progranulin mutation status in frontotemporal dementia patients and asymptomatic family members. Brain 132:583–591

    PubMed Central  PubMed  Google Scholar 

  10. Brouwers N, Nuytemans K, van der Zee J et al (2007) Alzheimer and Parkinson diagnoses in progranulin null mutation carriers in an extended founder family. Arch Neurol 64:1436–1446

    PubMed  Google Scholar 

  11. 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

    CAS  PubMed Central  PubMed  Google Scholar 

  12. Bertram L, McQueen MB, Mullin K et al (2007) Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat Genet 39:17–23

    CAS  PubMed  Google Scholar 

  13. Harold D, Abraham R, Hollingworth P et al (2009) Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat Genet 41:1088–1093

    CAS  PubMed Central  PubMed  Google Scholar 

  14. Lambert JC, Heath S, Even G et al (2009) Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat Genet 41:1094–1099

    CAS  PubMed  Google Scholar 

  15. Seshadri S, Fitzpatrick AL, Ikram MA et al (2010) Genome-wide analysis of genetic loci associated with Alzheimer disease. JAMA 303:1832–1840

    CAS  PubMed Central  PubMed  Google Scholar 

  16. Hollingworth P, Harold D, Sims R et al (2011) Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer’s disease. Nat Genet 43:429–435

    CAS  PubMed Central  PubMed  Google Scholar 

  17. Naj AC, Jun G, Beecham GW et al (2011) Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer’s disease. Nat Genet 43:436–441

    CAS  PubMed Central  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  19. Jones L, Holmans PA, Hamshere ML et al (2010) Genetic evidence implicates the immune system and cholesterol metabolism in the etiology of Alzheimer’s disease. PLoS One 5:e13950

    PubMed Central  PubMed  Google Scholar 

  20. Lambert JC, Ibrahim-Verbaas CA, Harold D et al (2013) Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet 45:1452–1458

    CAS  PubMed Central  PubMed  Google Scholar 

  21. Jonsson T, Stefansson H, Steinberg S et al (2013) Variant of TREM2 associated with the risk of Alzheimer’s disease. N Engl J Med 368:107–116

    CAS  PubMed Central  PubMed  Google Scholar 

  22. Guerreiro R, Wojtas A, Bras J et al (2013) TREM2 variants in Alzheimer’s disease. N Engl J Med 368:117–127

    CAS  PubMed Central  PubMed  Google Scholar 

  23. Neumann H, Daly MJ (2013) Variant TREM2 as risk factor for Alzheimer’s disease. N Engl J Med 368:182–184

    CAS  PubMed  Google Scholar 

  24. Chinn S (1989) Longitudinal studies: design and analysis. Rev Epidemiol Sante Publique 37:431–441

    CAS  PubMed  Google Scholar 

  25. Gibbons RD (2008) Design and analysis of longitudinal studies. Psychiatr Ann 38:758–761

    PubMed Central  PubMed  Google Scholar 

  26. Liu C, Cripe TP, Kim MO (2010) Statistical issues in longitudinal data analysis for treatment efficacy studies in the biomedical sciences. Mol Ther 18:1724–1730

    CAS  PubMed Central  PubMed  Google Scholar 

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

    PubMed  Google Scholar 

  28. 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

    CAS  PubMed  Google Scholar 

  29. 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

    CAS  PubMed  Google Scholar 

  30. Rosén C, Hansson O, Blennow K et al (2013) Fluid biomarkers in Alzheimer’s disease - current concepts. Mol Neurodegener 8:20

    PubMed Central  PubMed  Google Scholar 

  31. Carrillo MC, Blennow K, Soares H et al (2013) Global standardization measurement of cerebral spinal fluid for Alzheimer’s disease: an update from the Alzheimer’s Association Global Biomarkers Consortium. Alzheimers Dement 9:137–140

    PubMed  Google Scholar 

  32. Villemagne VL, Burnham S, Bourgeat P et al (2013) Amyloid β deposition, neurodegeneration, and cognitive decline in sporadic Alzheimer’s disease: a prospective cohort study. Lancet Neurol 12:357–367

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed Central  PubMed  Google Scholar 

  34. Noorbakhsh F, Overall CM, Power C (2009) Deciphering complex mechanisms in neurodegenerative diseases: the advent of systems biology. Trends Neurosci 32:88–100

    CAS  PubMed  Google Scholar 

  35. Tyers M, Mann M (2003) From genomics to proteomics. Nature 422:193–197

    CAS  PubMed  Google Scholar 

  36. Dong Z, Chen Y (2013) Transcriptomics: advances and approaches. Sci China Life Sci 56:960–967

    CAS  PubMed  Google Scholar 

  37. Wang Z, Gerstein M, Snyder M (2009) RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet 10:57–63

    CAS  PubMed Central  PubMed  Google Scholar 

  38. Liang WS, Dunckley T, Beach TG et al (2008) Altered neuronal gene expression in brain regions differentially affected by Alzheimer’s disease: a reference data set. Physiol Genomics 33:240–256

    CAS  PubMed Central  PubMed  Google Scholar 

  39. Tan MG, Chua WT, Esiri MM et al (2010) Genome wide profiling of altered gene expression in the neocortex of Alzheimer’s disease. J Neurosci Res 88:1157–1169

    CAS  PubMed  Google Scholar 

  40. Maes OC, Xu S, Yu B et al (2007) Transcriptional profiling of Alzheimer blood mononuclear cells by microarray. Neurobiol Aging 28:1795–1809

    CAS  PubMed  Google Scholar 

  41. Colangelo V, Schurr J, Ball MJ et al (2002) Gene expression profiling of 12633 genes in Alzheimer hippocampal CA1: transcription and neurotrophic factor down-regulation and upregulation of apoptotic and pro-inflammatory signaling. J Neurosci Res 70:462–473

    CAS  PubMed  Google Scholar 

  42. Blalock EM, Geddes JW, Chen KC et al (2004) Incipient Alzheimer’s disease: microarray correlation analyses reveal major transcriptional and tumor suppressor responses. Proc Natl Acad Sci U S A 101:2173–2178

    CAS  PubMed Central  PubMed  Google Scholar 

  43. Yao PJ, Zhu M, Pyun EI et al (2003) Defects in expression of genes related to synaptic vesicle trafficking in frontal cortex of Alzheimer’s disease. Neurobiol Dis 12:97–109

    CAS  PubMed  Google Scholar 

  44. Fehlbaum-Beurdeley P, Jarrige-Le Prado AC, Pallares D et al (2010) Toward an Alzheimer’s disease diagnosis via high-resolution blood gene expression. Alzheimers Dement 6:25–38

    CAS  PubMed  Google Scholar 

  45. Chen KD, Chang PT, Ping YH et al (2011) Gene expression profiling of peripheral blood leukocytes identifies and validates ABCB1 as a novel biomarker for Alzheimer’s disease. Neurobiol Dis 43:698–705

    CAS  PubMed  Google Scholar 

  46. Booij BB, Lindahl T, Wetterberg P et al (2011) A gene expression pattern in blood for the early detection of Alzheimer’s disease. J Alzheimers Dis 23:109–119

    CAS  PubMed  Google Scholar 

  47. Fehlbaum-Beurdeley P, Sol O, Désiré L et al (2012) Validation of AclarusDx™, a blood-based transcriptomic signature for the diagnosis of Alzheimer’s disease. J Alzheimers Dis 32:169–181

    CAS  PubMed  Google Scholar 

  48. Han G, Wang J, Zeng F et al (2013) Characteristic transformation of blood transcriptome in Alzheimer’s disease. J Alzheimers Dis 35:373–386

    CAS  PubMed  Google Scholar 

  49. Wasinger VC, Cordwell SJ, Cerpa-Poljak A et al (1995) Progress with gene-product mapping of the Mollicutes: Mycoplasma genitalium. Electrophoresis 16:1090–1094

    CAS  PubMed  Google Scholar 

  50. Tambor V, Fucíková A, Lenco J et al (2010) Application of proteomics in biomarker discovery: a primer for the clinician. Physiol Res 59:471–497

    CAS  PubMed  Google Scholar 

  51. Thambisetty M, Lovestone S (2010) Blood-based biomarkers of Alzheimer’s disease: challenging but feasible. Biomark Med 4:65–79

    CAS  PubMed Central  PubMed  Google Scholar 

  52. Becker M, Schindler J, Nothwang HG (2006) Neuroproteomics – the tasks lying ahead. Electrophoresis 27:2819–2829

    CAS  PubMed  Google Scholar 

  53. Hamacher M, Meyer HE (2005) HUPO Brain Proteome Project: aims and needs in proteomics. Exp Rev Proteomics 2:1–3

    Google Scholar 

  54. Rifai N, Gillette MA, Carr SA et al (2006) Protein biomarker discovery and validation: the long and uncertain path to clinical utility. Nat Biotechnol 24:971–983

    CAS  PubMed  Google Scholar 

  55. Portelius E, Gustavsson MK, Zetterberg H et al (2012) Evaluation of the performance of novel Aβ isoforms as theragnostic markers in Alzheimer’s disease: from the cell to the patient. Neurodegener Dis 10:138–140

    CAS  PubMed  Google Scholar 

  56. Portelius E, Price E, Brinkmalm G et al (2011) A novel pathway for amyloid precursor protein processing. Neurobiol Aging 32:1090–1098

    CAS  PubMed  Google Scholar 

  57. Perrin RJ, Craig-Schapiro R, Malone JP et al (2011) Identification and validation of novel cerebrospinal fluid biomarkers for staging early Alzheimer’s disease. PLoS One 6:e16032

    CAS  PubMed Central  PubMed  Google Scholar 

  58. Craig-Schapiro R, Kuhn M, Xiong C et al (2011) Multiplexed immunoassay panel identifies novel CSF biomarkers for Alzheimer’s disease diagnosis and prognosis. PLoS One 6:e18850

    CAS  PubMed Central  PubMed  Google Scholar 

  59. Craig-Schapiro R, Perrin RJ, Roe CM et al (2010) YKL-40: a novel prognostic fluid biomarker for preclinical Alzheimer’s disease. Biol Psychiatry 68:903–912

    CAS  PubMed Central  PubMed  Google Scholar 

  60. Portelius E, Dean RA, Gustavsson MK et al (2010) A novel Abeta isoform pattern in CSF reflects gamma-secretase inhibition in Alzheimer disease. Alzheimers Res Ther 2:7

    PubMed Central  PubMed  Google Scholar 

  61. Albertini V, Bruno A, Paterlini A et al (2010) Optimization protocol for amyloid-β peptides detection in human cerebrospinal fluid using SELDI TOF MS. Proteomics Clin Appl 4:352–357

    CAS  PubMed  Google Scholar 

  62. Portelius E, Brinkmalm G, Tran AJ et al (2009) Identification of novel APP/Abeta isoforms in human cerebrospinal fluid. Neurodegener Dis 6:87–94

    CAS  PubMed  Google Scholar 

  63. Simonsen AH, McGuire J, Podust VN et al (2008) Identification of a novel panel of cerebrospinal fluid biomarkers for Alzheimer’s disease. Neurobiol Aging 29:961–968

    CAS  PubMed  Google Scholar 

  64. Simonsen AH, McGuire J, Hansson O et al (2007) Novel panel of cerebrospinal fluid biomarkers for the prediction of progression to Alzheimer dementia in patients with mild cognitive impairment. Arch Neurol 64:366–370

    PubMed  Google Scholar 

  65. Simonsen AH, McGuire J, Podust VN et al (2007) A novel panel of cerebrospinal fluid biomarkers for the differential diagnosis of Alzheimer’s disease versus normal aging and frontotemporal dementia. Dement Geriatr Cogn Disord 24:434–440

    CAS  PubMed  Google Scholar 

  66. Finehout EJ, Franck Z, Choe LH et al (2007) Cerebrospinal fluid proteomic biomarkers for Alzheimer’s disease. Ann Neurol 61:120–129

    CAS  PubMed  Google Scholar 

  67. Portelius E, Tran AJ, Andreasson U et al (2007) Characterization of amyloid beta peptides in cerebrospinal fluid by an automated immunoprecipitation procedure followed by mass spectrometry. J Proteome Res 6:4433–4439

    CAS  PubMed  Google Scholar 

  68. Portelius E, Zetterberg H, Andreasson U et al (2006) An Alzheimer’s disease-specific beta amyloid fragment signature in cerebrospinal fluid. Neurosci Lett 409:215–219

    CAS  PubMed  Google Scholar 

  69. Portelius E, Westman-Brinkmalm A, Zetterberg H et al (2006) Determination of beta-amyloid peptide signatures in cerebrospinal fluid using immunoprecipitation–mass spectrometry. J Proteome Res 5:1010–1016

    CAS  PubMed  Google Scholar 

  70. Carrette O, Demalte I, Scherl A et al (2003) A panel of cerebrospinal fluid potential biomarkers for the diagnosis of Alzheimer’s disease. Proteomics 3:1486–1494

    CAS  PubMed  Google Scholar 

  71. Guo LH, Alexopoulos P, Wagenpfeil S (2013) Plasma proteomics for the identification of Alzheimer disease. Alzheimer Dis Assoc Disord. doi:10.1097/WAD.0b013e31827b60d2

    PubMed Central  PubMed  Google Scholar 

  72. Doecke JD, Laws SM, Faux NG et al (2012) Blood-based protein biomarkers for diagnosis of Alzheimer disease. Arch Neurol 69:1318–1325

    PubMed  Google Scholar 

  73. Hu WT, Holtzman DM, Fagan AM et al (2012) Plasma multianalyte profiling in mild cognitive impairment and Alzheimer disease. Neurology 79:897–905

    CAS  PubMed Central  PubMed  Google Scholar 

  74. Soares HD, Potter WZ, Pickering E et al (2012) Plasma biomarkers associated with the apolipoprotein E genotype and Alzheimer disease. Arch Neurol 69:1310–1317

    PubMed Central  PubMed  Google Scholar 

  75. Johnstone D, Milward EA, Berretta R et al (2012) Multivariate protein signatures of pre-clinical Alzheimer’s disease in the Alzheimer’s disease neuroimaging initiative (ADNI) plasma proteome dataset. PLoS One 7:e34341

    CAS  PubMed Central  PubMed  Google Scholar 

  76. O’Bryant SE, Xiao G, Barber R et al (2011) A blood-based algorithm for the detection of Alzheimer’s disease. Dement Geriatr Cogn Disord 32:55–62

    PubMed Central  PubMed  Google Scholar 

  77. O’Bryant SE, Xiao G, Barber R et al (2011) A blood-based screening tool for Alzheimer’s disease that spans serum and plasma: findings from TARC and ADNI. PLoS One 6:e28092

    PubMed Central  PubMed  Google Scholar 

  78. O’Bryant SE, Xiao G, Barber R et al (2010) A serum protein-based algorithm for the detection of Alzheimer disease. Arch Neurol 67:1077–1081

    PubMed Central  PubMed  Google Scholar 

  79. Ray S, Britschgi M, Herbert C et al (2007) Classification and prediction of clinical Alzheimer’s diagnosis based on plasma signaling proteins. Nat Med 13:1359–1362

    CAS  PubMed  Google Scholar 

  80. Hye A, Lynham S, Thambisetty M et al (2006) Proteome-based plasma biomarkers for Alzheimer’s disease. Brain 129:3042–3050

    CAS  PubMed  Google Scholar 

  81. Henriksen K, O’Bryant SE, Hampel H et al (2013) The future of blood-based biomarkers for Alzheimer’s disease. Alzheimers Dement. doi:10.1016/j.jalz.2013.01.013

    PubMed Central  PubMed  Google Scholar 

  82. Gupta VB, Sundaram R, Martins RN (2013) Multiplex biomarkers in blood. Alzheimers Res Ther 5:31

    PubMed Central  PubMed  Google Scholar 

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

    PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  85. Portelius E, Zetterberg H, Gobom J et al (2008) Targeted proteomics in Alzheimer’s disease: focus on amyloid-beta. Expert Rev Proteomics 5:225–237

    CAS  PubMed  Google Scholar 

  86. Davidsson P, Sjogren M (2005) The use of proteomics in biomarker discovery in neurodegenerative diseases. Dis Markers 21:81–92

    CAS  PubMed Central  PubMed  Google Scholar 

  87. Irizarry MC (2004) Biomarkers of Alzheimer disease in plasma. NeuroRx 1:226–234

    PubMed Central  PubMed  Google Scholar 

  88. Fagan AM, Perrin RJ (2012) Upcoming candidate cerebrospinal fluid biomarkers of Alzheimer’s disease. Biomark Med 6:455–476

    CAS  PubMed Central  PubMed  Google Scholar 

  89. Kaddurah-Daouk R, Kristal BS, Weinshilboum RM (2008) Metabolomics: a global biochemical approach to drug response and disease. Annu Rev Pharmacol Toxicol 48:653–683

    CAS  PubMed  Google Scholar 

  90. Kristal BS, Kaddurah-Daouk R et al (2007) Metabolomics: concept and potential neuroscience application. In: Lajtha A, Gibson G, Dienel G (eds) Handbook of neurochemistry and molecular neurobiology. Brain energetics. Integration of molecular and cellular processes, 3rd edn. Springer, New York, pp 889–912

    Google Scholar 

  91. Sun J, Beger RD, Schnackenberg LK (2013) Metabolomics as a tool for personalizing medicine: 2012 update. Personal Med 10:149–161

    CAS  Google Scholar 

  92. Smith CA, Want EJ, O’Maille G et al (2006) XCMS: processing mass spectrometry data for metabolite profiling using nonlinear peak alignment, matching, and identification. Anal Chem 78:779–787

    CAS  PubMed  Google Scholar 

  93. Baker M (2011) Metabolomics: from small molecules to big ideas. Nat Methods 8:117–121

    CAS  Google Scholar 

  94. Trushina E, Dutta T, Persson XM et al (2013) Identification of altered metabolic pathways in plasma and CSF in mild cognitive impairment and Alzheimer’s disease using metabolomics. PLoS One 8:e63644

    CAS  PubMed Central  PubMed  Google Scholar 

  95. Han X, MHoltzman D, McKeel DW Jr (2002) Substantial sulfatide deficiency and ceramide elevation in very early Alzheimer’s disease: potential role in disease pathogenesis. J Neurochem 82:809–818

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  97. Dunn WB, Broadhurst D, Begley P et al (2011) Procedures for large-scale metabolic profiling of serum and plasma using gas chromatography and liquid chromatography coupled to mass spectrometry. Nat Protoc 6:1060–1083

    CAS  PubMed  Google Scholar 

  98. Cottingham K (2008) HUSERMET researchers look to the metabolome for answers. J Proteome Res 7:4213

    Google Scholar 

  99. Han X (2010) Multi-dimensional mass spectrometry-based shotgun lipidomics and the altered lipids at the mild cognitive impairment stage of Alzheimer’s disease. Biochim Biophys Acta 1801:774–783

    CAS  PubMed Central  PubMed  Google Scholar 

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

    CAS  PubMed Central  PubMed  Google Scholar 

  101. Czech C, Berndt P, Busch K et al (2012) Metabolite profiling of Alzheimer’s disease cerebrospinal fluid. PLoS One 7:e31501

    CAS  PubMed Central  PubMed  Google Scholar 

  102. Kaddurah-Daouk R, Rozen S, Matson W et al (2011) Metabolomic changes in autopsy-confirmed Alzheimer’s disease. Alzheimers Dement 7:309–317

    PubMed Central  PubMed  Google Scholar 

  103. Sato Y, Suzuki I, Nakamura T et al (2012) Identification of a new plasma biomarker of Alzheimer’s disease using metabolomics technology. J Lipid Res 53:567–576

    CAS  PubMed Central  PubMed  Google Scholar 

  104. Orešič M, Hyötyläinen T, Herukka SK et al (2011) Metabolome in progression to Alzheimer’s disease. Transl Psychiatry 1:e57

    PubMed Central  PubMed  Google Scholar 

  105. Han X, Rozen S, Boyle SH et al (2011) Metabolomics in early Alzheimer’s disease: identification of altered plasma sphingolipidome using shotgun lipidomics. PLoS One 6:e21643

    CAS  PubMed Central  PubMed  Google Scholar 

  106. Wenk MR (2010) Lipidomics: new tools and applications. Cell 143:888–895

    CAS  PubMed  Google Scholar 

  107. Astarita G, Piomelli D (2011) Towards a whole-body systems [multi-organ] lipidomics in Alzheimer’s disease. Prostaglandins Leukot Essent Fatty Acids 85:197–203

    CAS  PubMed Central  PubMed  Google Scholar 

  108. Niemelä PS, Ollila S, Hyvönen MT et al (2007) Assessing the nature of lipid raft membranes. PLoS Comput Biol 3:e34

    PubMed Central  PubMed  Google Scholar 

  109. Yetukuri L, Söderlund S, Koivuniemi A et al (2010) Composition and lipid spatial distribution of HDL particles in subjects with low and high HDL-cholesterol. J Lipid Res 51:2341–2351

    CAS  PubMed Central  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  111. Horvath S, Dong J (2008) Geometric interpretation of gene coexpression network analysis. PLoS Comput Biol 4:e1000117

    PubMed Central  PubMed  Google Scholar 

  112. Langfelder P, Horvath S (2008) WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9:559

    PubMed Central  PubMed  Google Scholar 

  113. Langfelder P, Horvath S (2007) Eigengene networks for studying the relationships between co-expression modules. BMC Syst Biol 1:54

    PubMed Central  PubMed  Google Scholar 

  114. Miller JA, Oldham MC, Geschwind DH (2008) A systems level analysis of transcriptional changes in Alzheimer’s disease and normal aging. J Neurosci 28:1410–1420

    CAS  PubMed Central  PubMed  Google Scholar 

  115. Miller JA, Horvath S, Geschwind DH (2010) Divergence of human and mouse brain transcriptome highlights Alzheimer disease pathways. Proc Natl Acad Sci U S A 107:12698–12703

    CAS  PubMed Central  PubMed  Google Scholar 

  116. Kann MG (2007) Protein interactions and disease: computational approaches to uncover the etiology of diseases. Brief Bioinform 8:333–346

    CAS  PubMed  Google Scholar 

  117. Barabasi AL, Oltvai ZN (2004) Network biology: understanding the cell’s functional organization. Nat Rev Genet 5:101–115

    CAS  PubMed  Google Scholar 

  118. Hallock P, Thomas MA (2012) Integrating the Alzheimer’s disease proteome and transcriptome: a comprehensive network model of a complex disease. OMICS 16:37–49

    CAS  PubMed Central  PubMed  Google Scholar 

  119. Prasad TSK, Goel R, Kandasamy K et al (2009) Human Protein Reference Database-2009 update. Nucleic Acids Res 37:D767–D772

    CAS  Google Scholar 

  120. Orešič M, Lötjönen J, Soininen H (2010) Systems medicine and the integration of bioinformatic tools for the diagnosis of Alzheimer’s disease. Genome Med 2:83

    PubMed Central  Google Scholar 

Download references

Acknowledgements

HH and BD would like to thank for the support of the Fondation Pour La Recherche Sur Alzheimer (FRA), Paris, France.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Simone Lista Ph.D. .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2016 Springer Science+Business Media New York

About this protocol

Cite this protocol

Lista, S., Khachaturian, Z.S., Rujescu, D., Garaci, F., Dubois, B., Hampel, H. (2016). Application of Systems Theory in Longitudinal Studies on the Origin and Progression of Alzheimer’s Disease. In: Castrillo, J., Oliver, S. (eds) Systems Biology of Alzheimer's Disease. Methods in Molecular Biology, vol 1303. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-2627-5_2

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-2627-5_2

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-2626-8

  • Online ISBN: 978-1-4939-2627-5

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics