Skip to main content
Top
Published in: Alzheimer's Research & Therapy 1/2017

Open Access 01-12-2017 | Research

Analyzing the genes related to Alzheimer’s disease via a network and pathway-based approach

Authors: Yan-Shi Hu, Juncai Xin, Ying Hu, Lei Zhang, Ju Wang

Published in: Alzheimer's Research & Therapy | Issue 1/2017

Login to get access

Abstract

Background

Our understanding of the molecular mechanisms underlying Alzheimer’s disease (AD) remains incomplete. Previous studies have revealed that genetic factors provide a significant contribution to the pathogenesis and development of AD. In the past years, numerous genes implicated in this disease have been identified via genetic association studies on candidate genes or at the genome-wide level. However, in many cases, the roles of these genes and their interactions in AD are still unclear. A comprehensive and systematic analysis focusing on the biological function and interactions of these genes in the context of AD will therefore provide valuable insights to understand the molecular features of the disease.

Method

In this study, we collected genes potentially associated with AD by screening publications on genetic association studies deposited in PubMed. The major biological themes linked with these genes were then revealed by function and biochemical pathway enrichment analysis, and the relation between the pathways was explored by pathway crosstalk analysis. Furthermore, the network features of these AD-related genes were analyzed in the context of human interactome and an AD-specific network was inferred using the Steiner minimal tree algorithm.

Results

We compiled 430 human genes reported to be associated with AD from 823 publications. Biological theme analysis indicated that the biological processes and biochemical pathways related to neurodevelopment, metabolism, cell growth and/or survival, and immunology were enriched in these genes. Pathway crosstalk analysis then revealed that the significantly enriched pathways could be grouped into three interlinked modules—neuronal and metabolic module, cell growth/survival and neuroendocrine pathway module, and immune response-related module—indicating an AD-specific immune-endocrine-neuronal regulatory network. Furthermore, an AD-specific protein network was inferred and novel genes potentially associated with AD were identified.

Conclusion

By means of network and pathway-based methodology, we explored the pathogenetic mechanism underlying AD at a systems biology level. Results from our work could provide valuable clues for understanding the molecular mechanism underlying AD. In addition, the framework proposed in this study could be used to investigate the pathological molecular network and genes relevant to other complex diseases or phenotypes.
Appendix
Available only for authorised users
Literature
1.
go back to reference Ballard C, Gauthier S, Corbett A, Brayne C, Aarsland D, Jones E. Alzheimer’s disease. Lancet. 2011;377(9770):1019–31.CrossRefPubMed Ballard C, Gauthier S, Corbett A, Brayne C, Aarsland D, Jones E. Alzheimer’s disease. Lancet. 2011;377(9770):1019–31.CrossRefPubMed
3.
go back to reference Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, Adair T, Aggarwal R, Ahn SY, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380(9859):2095–128.CrossRefPubMed Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, Abraham J, Adair T, Aggarwal R, Ahn SY, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380(9859):2095–128.CrossRefPubMed
5.
go back to reference Ager RR, Davis JL, Agazaryan A, Benavente F, Poon WW, LaFerla FM, Blurton-Jones M. Human neural stem cells improve cognition and promote synaptic growth in two complementary transgenic models of Alzheimer’s disease and neuronal loss. Hippocampus. 2015;25(7):813–26.CrossRefPubMedPubMedCentral Ager RR, Davis JL, Agazaryan A, Benavente F, Poon WW, LaFerla FM, Blurton-Jones M. Human neural stem cells improve cognition and promote synaptic growth in two complementary transgenic models of Alzheimer’s disease and neuronal loss. Hippocampus. 2015;25(7):813–26.CrossRefPubMedPubMedCentral
6.
go back to reference Bateman RJ, Xiong C, Benzinger TL, Fagan AM, Goate A, Fox NC, Marcus DS, Cairns NJ, Xie X, Blazey TM, et al. Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. N Engl J Med. 2012;367(9):795–804.CrossRefPubMedPubMedCentral Bateman RJ, Xiong C, Benzinger TL, Fagan AM, Goate A, Fox NC, Marcus DS, Cairns NJ, Xie X, Blazey TM, et al. Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. N Engl J Med. 2012;367(9):795–804.CrossRefPubMedPubMedCentral
7.
go back to reference Solomon A, Mangialasche F, Richard E, Andrieu S, Bennett DA, Breteler M, Fratiglioni L, Hooshmand B, Khachaturian AS, Schneider LS, et al. Advances in the prevention of Alzheimer’s disease and dementia. J Intern Med. 2014;275(3):229–50.CrossRefPubMedPubMedCentral Solomon A, Mangialasche F, Richard E, Andrieu S, Bennett DA, Breteler M, Fratiglioni L, Hooshmand B, Khachaturian AS, Schneider LS, et al. Advances in the prevention of Alzheimer’s disease and dementia. J Intern Med. 2014;275(3):229–50.CrossRefPubMedPubMedCentral
8.
go back to reference Ryu JK, Cho T, Choi HB, Jantaratnotai N, McLarnon JG. Pharmacological antagonism of interleukin-8 receptor CXCR2 inhibits inflammatory reactivity and is neuroprotective in an animal model of Alzheimer’s disease. J Neuroinflammation. 2015;12:144.CrossRefPubMedPubMedCentral Ryu JK, Cho T, Choi HB, Jantaratnotai N, McLarnon JG. Pharmacological antagonism of interleukin-8 receptor CXCR2 inhibits inflammatory reactivity and is neuroprotective in an animal model of Alzheimer’s disease. J Neuroinflammation. 2015;12:144.CrossRefPubMedPubMedCentral
9.
go back to reference Allen M, Zou F, Chai HS, Younkin CS, Crook J, Pankratz VS, Carrasquillo MM, Rowley CN, Nair AA, Middha S, et al. Novel late-onset Alzheimer disease loci variants associate with brain gene expression. Neurology. 2012;79(3):221–8.CrossRefPubMedPubMedCentral Allen M, Zou F, Chai HS, Younkin CS, Crook J, Pankratz VS, Carrasquillo MM, Rowley CN, Nair AA, Middha S, et al. Novel late-onset Alzheimer disease loci variants associate with brain gene expression. Neurology. 2012;79(3):221–8.CrossRefPubMedPubMedCentral
10.
go back to reference Naj AC, Jun G, Reitz C, Kunkle BW, Perry W, Park YS, Beecham GW, Rajbhandary RA, Hamilton-Nelson KL, Wang LS, et al. Effects of multiple genetic loci on age at onset in late-onset Alzheimer disease: a genome-wide association study. JAMA Neurol. 2014;71(11):1394–404.CrossRefPubMedPubMedCentral Naj AC, Jun G, Reitz C, Kunkle BW, Perry W, Park YS, Beecham GW, Rajbhandary RA, Hamilton-Nelson KL, Wang LS, et al. Effects of multiple genetic loci on age at onset in late-onset Alzheimer disease: a genome-wide association study. JAMA Neurol. 2014;71(11):1394–404.CrossRefPubMedPubMedCentral
11.
go back to reference Cabral C, Morgado PM, Campos Costa D, Silveira M. Alzheimers Disease Neuroimaging Initiative. Predicting conversion from MCI to AD with FDG-PET brain images at different prodromal stages. Comput Biol Med. 2015;58:101–9.CrossRefPubMed Cabral C, Morgado PM, Campos Costa D, Silveira M. Alzheimers Disease Neuroimaging Initiative. Predicting conversion from MCI to AD with FDG-PET brain images at different prodromal stages. Comput Biol Med. 2015;58:101–9.CrossRefPubMed
12.
go back to reference Gatz M, Reynolds CA, Fratiglioni L, Johansson B, Mortimer JA, Berg S, Fiske A, Pedersen NL. Role of genes and environments for explaining Alzheimer disease. Arch Gen Psychiatry. 2006;63(2):168–74.CrossRefPubMed Gatz M, Reynolds CA, Fratiglioni L, Johansson B, Mortimer JA, Berg S, Fiske A, Pedersen NL. Role of genes and environments for explaining Alzheimer disease. Arch Gen Psychiatry. 2006;63(2):168–74.CrossRefPubMed
14.
go back to reference Zhang B, Kirov S, Snoddy J. WebGestalt: an integrated system for exploring gene sets in various biological contexts. Nucleic Acids Res. 2005;33(Web Server issue):W741–8.CrossRefPubMedPubMedCentral Zhang B, Kirov S, Snoddy J. WebGestalt: an integrated system for exploring gene sets in various biological contexts. Nucleic Acids Res. 2005;33(Web Server issue):W741–8.CrossRefPubMedPubMedCentral
15.
go back to reference Chen J, Bardes EE, Aronow BJ, Jegga AG. ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. 2009;37(Web Server issue):W305–11.CrossRefPubMedPubMedCentral Chen J, Bardes EE, Aronow BJ, Jegga AG. ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. 2009;37(Web Server issue):W305–11.CrossRefPubMedPubMedCentral
16.
go back to reference Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.CrossRefPubMedPubMedCentral Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.CrossRefPubMedPubMedCentral
17.
go back to reference Cowley MJ, Pinese M, Kassahn KS, Waddell N, Pearson JV, Grimmond SM, Biankin AV, Hautaniemi S, Wu J. PINA v2.0: mining interactome modules. Nucleic Acids Res. 2012;40(Database issue):D862–5.CrossRefPubMed Cowley MJ, Pinese M, Kassahn KS, Waddell N, Pearson JV, Grimmond SM, Biankin AV, Hautaniemi S, Wu J. PINA v2.0: mining interactome modules. Nucleic Acids Res. 2012;40(Database issue):D862–5.CrossRefPubMed
18.
go back to reference Menche J, Sharma A, Kitsak M, Ghiassian SD, Vidal M, Loscalzo J, Barabasi AL. Disease networks. Uncovering disease-disease relationships through the incomplete interactome. Science. 2015;347(6224):1257601.CrossRefPubMedPubMedCentral Menche J, Sharma A, Kitsak M, Ghiassian SD, Vidal M, Loscalzo J, Barabasi AL. Disease networks. Uncovering disease-disease relationships through the incomplete interactome. Science. 2015;347(6224):1257601.CrossRefPubMedPubMedCentral
20.
go back to reference Klein P, Ravi R. A nearly best-possible approximation algorithm for node-weighted Steiner trees. J Algorithms. 1995;19(1):104–15.CrossRef Klein P, Ravi R. A nearly best-possible approximation algorithm for node-weighted Steiner trees. J Algorithms. 1995;19(1):104–15.CrossRef
21.
go back to reference Zheng S, Zhao Z. GenRev: exploring functional relevance of genes in molecular networks. Genomics. 2012;99(3):183–8.CrossRefPubMed Zheng S, Zhao Z. GenRev: exploring functional relevance of genes in molecular networks. Genomics. 2012;99(3):183–8.CrossRefPubMed
22.
go back to reference Erdos P, Rényi A. On the evolution of random graphs. Publ Math Inst Hungar Acad Sci. 1960;5:17–61. Erdos P, Rényi A. On the evolution of random graphs. Publ Math Inst Hungar Acad Sci. 1960;5:17–61.
23.
go back to reference Roussotte FF, Daianu M, Jahanshad N, Leonardo CD, Thompson PM. Neuroimaging and genetic risk for Alzheimer’s disease and addiction-related degenerative brain disorders. Brain Imaging Behav. 2014;8(2):217–33.CrossRefPubMedPubMedCentral Roussotte FF, Daianu M, Jahanshad N, Leonardo CD, Thompson PM. Neuroimaging and genetic risk for Alzheimer’s disease and addiction-related degenerative brain disorders. Brain Imaging Behav. 2014;8(2):217–33.CrossRefPubMedPubMedCentral
24.
go back to reference Anstey KJ, Mack HA, Cherbuin N. Alcohol consumption as a risk factor for dementia and cognitive decline: meta-analysis of prospective studies. Am J Geriatr Psychiatry. 2009;17(7):542–55.CrossRefPubMed Anstey KJ, Mack HA, Cherbuin N. Alcohol consumption as a risk factor for dementia and cognitive decline: meta-analysis of prospective studies. Am J Geriatr Psychiatry. 2009;17(7):542–55.CrossRefPubMed
25.
go back to reference Gjoneska E, Pfenning AR, Mathys H, Quon G, Kundaje A, Tsai LH, Kellis M. Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer’s disease. Nature. 2015;518(7539):365–9.CrossRefPubMedPubMedCentral Gjoneska E, Pfenning AR, Mathys H, Quon G, Kundaje A, Tsai LH, Kellis M. Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer’s disease. Nature. 2015;518(7539):365–9.CrossRefPubMedPubMedCentral
26.
go back to reference Heneka MT, Golenbock DT, Latz E. Innate immunity in Alzheimer’s disease. Nat Immunol. 2015;16(3):229–36.CrossRefPubMed Heneka MT, Golenbock DT, Latz E. Innate immunity in Alzheimer’s disease. Nat Immunol. 2015;16(3):229–36.CrossRefPubMed
27.
go back to reference Wang C, Zhang X, Teng Z, Zhang T, Li Y. Downregulation of PI3K/Akt/mTOR signaling pathway in curcumin-induced autophagy in APP/PS1 double transgenic mice. Eur J Pharmacol. 2014;740:312–20.CrossRefPubMed Wang C, Zhang X, Teng Z, Zhang T, Li Y. Downregulation of PI3K/Akt/mTOR signaling pathway in curcumin-induced autophagy in APP/PS1 double transgenic mice. Eur J Pharmacol. 2014;740:312–20.CrossRefPubMed
28.
go back to reference Polychronidou E, Vlachakis D, Vlamos P, Baumann M, Kossida S. Notch signaling and ageing. Adv Exp Med Biol. 2015;822:25–36.CrossRefPubMed Polychronidou E, Vlachakis D, Vlamos P, Baumann M, Kossida S. Notch signaling and ageing. Adv Exp Med Biol. 2015;822:25–36.CrossRefPubMed
29.
go back to reference Wang D, Ho L, Faith J, Ono K, Janle EM, Lachcik PJ, Cooper BR, Jannasch AH, D’Arcy BR, Williams BA, et al. Role of intestinal microbiota in the generation of polyphenol-derived phenolic acid mediated attenuation of Alzheimer’s disease beta-amyloid oligomerization. Mol Nutr Food Res. 2015;59(6):1025–40.CrossRefPubMedPubMedCentral Wang D, Ho L, Faith J, Ono K, Janle EM, Lachcik PJ, Cooper BR, Jannasch AH, D’Arcy BR, Williams BA, et al. Role of intestinal microbiota in the generation of polyphenol-derived phenolic acid mediated attenuation of Alzheimer’s disease beta-amyloid oligomerization. Mol Nutr Food Res. 2015;59(6):1025–40.CrossRefPubMedPubMedCentral
30.
go back to reference Alam MZ, Alam Q, Kamal MA, Abuzenadah AM, Haque A. A possible link of gut microbiota alteration in type 2 diabetes and Alzheimer’s disease pathogenicity: an update. CNS Neurol Disord Drug Targets. 2014;13(3):383–90.CrossRefPubMed Alam MZ, Alam Q, Kamal MA, Abuzenadah AM, Haque A. A possible link of gut microbiota alteration in type 2 diabetes and Alzheimer’s disease pathogenicity: an update. CNS Neurol Disord Drug Targets. 2014;13(3):383–90.CrossRefPubMed
31.
32.
go back to reference Letra L, Santana I, Seica R. Obesity as a risk factor for Alzheimer’s disease: the role of adipocytokines. Metab Brain Dis. 2014;29(3):563–8.CrossRefPubMed Letra L, Santana I, Seica R. Obesity as a risk factor for Alzheimer’s disease: the role of adipocytokines. Metab Brain Dis. 2014;29(3):563–8.CrossRefPubMed
33.
go back to reference Teixeira AL, Diniz BS, Campos AC, Miranda AS, Rocha NP, Talib LL, Gattaz WF, Forlenza OV. Decreased levels of circulating adiponectin in mild cognitive impairment and Alzheimer’s disease. Neruomol Med. 2013;15(1):115–21.CrossRef Teixeira AL, Diniz BS, Campos AC, Miranda AS, Rocha NP, Talib LL, Gattaz WF, Forlenza OV. Decreased levels of circulating adiponectin in mild cognitive impairment and Alzheimer’s disease. Neruomol Med. 2013;15(1):115–21.CrossRef
34.
go back to reference Jia P, Kao CF, Kuo PH, Zhao Z. A comprehensive network and pathway analysis of candidate genes in major depressive disorder. BMC Syst Biol. 2011;5 Suppl 3:S12.CrossRefPubMedPubMedCentral Jia P, Kao CF, Kuo PH, Zhao Z. A comprehensive network and pathway analysis of candidate genes in major depressive disorder. BMC Syst Biol. 2011;5 Suppl 3:S12.CrossRefPubMedPubMedCentral
35.
go back to reference Leal MC, Magnani N, Villordo S, Buslje CM, Evelson P, Castano EM, Morelli L. Transcriptional regulation of insulin-degrading enzyme modulates mitochondrial amyloid beta (Abeta) peptide catabolism and functionality. J Biol Chem. 2013;288(18):12920–31.CrossRefPubMedPubMedCentral Leal MC, Magnani N, Villordo S, Buslje CM, Evelson P, Castano EM, Morelli L. Transcriptional regulation of insulin-degrading enzyme modulates mitochondrial amyloid beta (Abeta) peptide catabolism and functionality. J Biol Chem. 2013;288(18):12920–31.CrossRefPubMedPubMedCentral
36.
go back to reference Conejero-Goldberg C, Hyde TM, Chen S, Dreses-Werringloer U, Herman MM, Kleinman JE, Davies P, Goldberg TE. Molecular signatures in post-mortem brain tissue of younger individuals at high risk for Alzheimer’s disease as based on APOE genotype. Mol Psychiatry. 2011;16(8):836–47.CrossRefPubMed Conejero-Goldberg C, Hyde TM, Chen S, Dreses-Werringloer U, Herman MM, Kleinman JE, Davies P, Goldberg TE. Molecular signatures in post-mortem brain tissue of younger individuals at high risk for Alzheimer’s disease as based on APOE genotype. Mol Psychiatry. 2011;16(8):836–47.CrossRefPubMed
37.
go back to reference Adori C, Gluck L, Barde S, Yoshitake T, Kovacs GG, Mulder J, Magloczky Z, Havas L, Bolcskei K, Mitsios N, et al. Critical role of somatostatin receptor 2 in the vulnerability of the central noradrenergic system: new aspects on Alzheimer’s disease. Acta Neuropathol. 2015;129(4):541–63.CrossRefPubMed Adori C, Gluck L, Barde S, Yoshitake T, Kovacs GG, Mulder J, Magloczky Z, Havas L, Bolcskei K, Mitsios N, et al. Critical role of somatostatin receptor 2 in the vulnerability of the central noradrenergic system: new aspects on Alzheimer’s disease. Acta Neuropathol. 2015;129(4):541–63.CrossRefPubMed
38.
go back to reference Capsoni S, Amato G, Vignone D, Criscuolo C, Nykjaer A, Cattaneo A. Dissecting the role of sortilin receptor signaling in neurodegeneration induced by NGF deprivation. Biochem Biophys Res Commun. 2013;431(3):579–85.CrossRefPubMedPubMedCentral Capsoni S, Amato G, Vignone D, Criscuolo C, Nykjaer A, Cattaneo A. Dissecting the role of sortilin receptor signaling in neurodegeneration induced by NGF deprivation. Biochem Biophys Res Commun. 2013;431(3):579–85.CrossRefPubMedPubMedCentral
39.
go back to reference Satoh K, Abe-Dohmae S, Yokoyama S, St George-Hyslop P, Fraser PE. ATP-binding cassette transporter A7 (ABCA7) loss of function alters Alzheimer amyloid processing. J Biol Chem. 2015;290(40):24152–65.CrossRefPubMedPubMedCentral Satoh K, Abe-Dohmae S, Yokoyama S, St George-Hyslop P, Fraser PE. ATP-binding cassette transporter A7 (ABCA7) loss of function alters Alzheimer amyloid processing. J Biol Chem. 2015;290(40):24152–65.CrossRefPubMedPubMedCentral
40.
go back to reference Inkster B, Rao AW, Ridler K, Filippini N, Whitcher B, Nichols TE, Wetten S, Gibson RA, Borrie M, Kertesz A, et al. Genetic variation in GOLM1 and prefrontal cortical volume in Alzheimer’s disease. Neurobiol Aging. 2012;33(3):457–65.CrossRefPubMed Inkster B, Rao AW, Ridler K, Filippini N, Whitcher B, Nichols TE, Wetten S, Gibson RA, Borrie M, Kertesz A, et al. Genetic variation in GOLM1 and prefrontal cortical volume in Alzheimer’s disease. Neurobiol Aging. 2012;33(3):457–65.CrossRefPubMed
41.
go back to reference Raha AA, Vaishnav RA, Friedland RP, Bomford A, Raha-Chowdhury R. The systemic iron-regulatory proteins hepcidin and ferroportin are reduced in the brain in Alzheimer’s disease. Acta Neuropathol Commun. 2013;1:55.CrossRefPubMedPubMedCentral Raha AA, Vaishnav RA, Friedland RP, Bomford A, Raha-Chowdhury R. The systemic iron-regulatory proteins hepcidin and ferroportin are reduced in the brain in Alzheimer’s disease. Acta Neuropathol Commun. 2013;1:55.CrossRefPubMedPubMedCentral
42.
go back to reference Kong W, Zhang J, Mou X, Yang Y. Integrating gene expression and protein interaction data for signaling pathway prediction of Alzheimer’s disease. Comput Math Methods Med. 2014;2014:340758.PubMedPubMedCentral Kong W, Zhang J, Mou X, Yang Y. Integrating gene expression and protein interaction data for signaling pathway prediction of Alzheimer’s disease. Comput Math Methods Med. 2014;2014:340758.PubMedPubMedCentral
43.
go back to reference Ponzoni I, Nueda M, Tarazona S, Gotz S, Montaner D, Dussaut J, Dopazo J, Conesa A. Pathway network inference from gene expression data. BMC Syst Biol. 2014;8 Suppl 2:S7.CrossRefPubMedPubMedCentral Ponzoni I, Nueda M, Tarazona S, Gotz S, Montaner D, Dussaut J, Dopazo J, Conesa A. Pathway network inference from gene expression data. BMC Syst Biol. 2014;8 Suppl 2:S7.CrossRefPubMedPubMedCentral
44.
go back to reference Sun Y, Bresell A, Rantalainen M, Hoglund K, Lebouvier T, Salter H. Alzheimer Disease Neuroimaging Initiative. An integrated bioinformatics approach for identifying genetic markers that predict cerebrospinal fluid biomarker p-tau181/Abeta1-42 ratio in ApoE4-negative mild cognitive impairment patients. J Alzheimers Dis. 2015;45(4):1061–76.PubMed Sun Y, Bresell A, Rantalainen M, Hoglund K, Lebouvier T, Salter H. Alzheimer Disease Neuroimaging Initiative. An integrated bioinformatics approach for identifying genetic markers that predict cerebrospinal fluid biomarker p-tau181/Abeta1-42 ratio in ApoE4-negative mild cognitive impairment patients. J Alzheimers Dis. 2015;45(4):1061–76.PubMed
45.
go back to reference Williams-Skipp C, Raman T, Valuck RJ, Watkins H, Palmer BE, Scheinman RI. Unmasking of a protective tumor necrosis factor receptor I-mediated signal in the collagen-induced arthritis model. Arthritis Rheum. 2009;60(2):408–18.CrossRefPubMedPubMedCentral Williams-Skipp C, Raman T, Valuck RJ, Watkins H, Palmer BE, Scheinman RI. Unmasking of a protective tumor necrosis factor receptor I-mediated signal in the collagen-induced arthritis model. Arthritis Rheum. 2009;60(2):408–18.CrossRefPubMedPubMedCentral
46.
go back to reference Parra MA, Saarimaki H, Bastin ME, Londono AC, Pettit L, Lopera F, Della Sala S, Abrahams S. Memory binding and white matter integrity in familial Alzheimer’s disease. Brain. 2015;138(Pt 5):1355–69.CrossRefPubMed Parra MA, Saarimaki H, Bastin ME, Londono AC, Pettit L, Lopera F, Della Sala S, Abrahams S. Memory binding and white matter integrity in familial Alzheimer’s disease. Brain. 2015;138(Pt 5):1355–69.CrossRefPubMed
47.
go back to reference Ahmadian-Attari MM, Dargahi L, Mosaddegh M, Kamalinejad M, Khallaghi B, Noorbala F, Ahmadiani A. Impairment of rat spatial learning and memory in a new model of cold water-induced chronic hypothermia: implication for Alzheimer’s disease. Neurotox Res. 2015;28(2):95–107.CrossRefPubMed Ahmadian-Attari MM, Dargahi L, Mosaddegh M, Kamalinejad M, Khallaghi B, Noorbala F, Ahmadiani A. Impairment of rat spatial learning and memory in a new model of cold water-induced chronic hypothermia: implication for Alzheimer’s disease. Neurotox Res. 2015;28(2):95–107.CrossRefPubMed
48.
go back to reference Peter-Derex L, Yammine P, Bastuji H, Croisile B. Sleep and Alzheimer’s disease. Sleep Med Rev. 2015;19:29–38.CrossRefPubMed Peter-Derex L, Yammine P, Bastuji H, Croisile B. Sleep and Alzheimer’s disease. Sleep Med Rev. 2015;19:29–38.CrossRefPubMed
49.
go back to reference Suzuki C, Yokote Y, Takahashi T. Changes in daily cognition and behavior of Alzheimer’s patients over time: a three-year evaluation using a daily cognition and behavior for Alzheimer’s disease scale. Dementia. 2015;14(1):126–35.CrossRefPubMed Suzuki C, Yokote Y, Takahashi T. Changes in daily cognition and behavior of Alzheimer’s patients over time: a three-year evaluation using a daily cognition and behavior for Alzheimer’s disease scale. Dementia. 2015;14(1):126–35.CrossRefPubMed
50.
go back to reference Satoh J, Tabunoki H, Ishida T, Saito Y, Arima K. Accumulation of a repulsive axonal guidance molecule RGMa in amyloid plaques: a possible hallmark of regenerative failure in Alzheimer’s disease brains. Neuropathol Appl Neurobiol. 2013;39(2):109–20.CrossRefPubMed Satoh J, Tabunoki H, Ishida T, Saito Y, Arima K. Accumulation of a repulsive axonal guidance molecule RGMa in amyloid plaques: a possible hallmark of regenerative failure in Alzheimer’s disease brains. Neuropathol Appl Neurobiol. 2013;39(2):109–20.CrossRefPubMed
51.
go back to reference Landlinger C, Oberleitner L, Gruber P, Noiges B, Yatsyk K, Santic R, Mandler M, Staffler G. Active immunization against complement factor C5a: a new therapeutic approach for Alzheimer’s disease. J Neuroinflammation. 2015;12:150.CrossRefPubMedPubMedCentral Landlinger C, Oberleitner L, Gruber P, Noiges B, Yatsyk K, Santic R, Mandler M, Staffler G. Active immunization against complement factor C5a: a new therapeutic approach for Alzheimer’s disease. J Neuroinflammation. 2015;12:150.CrossRefPubMedPubMedCentral
52.
go back to reference Alcolea D, Martinez-Lage P, Sanchez-Juan P, Olazaran J, Antunez C, Izagirre A, Ecay-Torres M, Estanga A, Clerigue M, Guisasola MC, et al. Amyloid precursor protein metabolism and inflammation markers in preclinical Alzheimer disease. Neurology. 2015;85(7):626–33.CrossRefPubMed Alcolea D, Martinez-Lage P, Sanchez-Juan P, Olazaran J, Antunez C, Izagirre A, Ecay-Torres M, Estanga A, Clerigue M, Guisasola MC, et al. Amyloid precursor protein metabolism and inflammation markers in preclinical Alzheimer disease. Neurology. 2015;85(7):626–33.CrossRefPubMed
53.
go back to reference Wang X, Hu X, Yang Y, Takata T, Sakurai T. Systemic pyruvate administration markedly reduces neuronal death and cognitive impairment in a rat model of Alzheimer’s disease. Exp Neurol. 2015;271:145–54.CrossRefPubMed Wang X, Hu X, Yang Y, Takata T, Sakurai T. Systemic pyruvate administration markedly reduces neuronal death and cognitive impairment in a rat model of Alzheimer’s disease. Exp Neurol. 2015;271:145–54.CrossRefPubMed
54.
go back to reference Ahmed T, Blum D, Burnouf S, Demeyer D, Buee-Scherrer V, D’Hooge R, Buee L, Balschun D. Rescue of impaired late-phase long-term depression in a tau transgenic mouse model. Neurobiol Aging. 2015;36(2):730–9.CrossRefPubMed Ahmed T, Blum D, Burnouf S, Demeyer D, Buee-Scherrer V, D’Hooge R, Buee L, Balschun D. Rescue of impaired late-phase long-term depression in a tau transgenic mouse model. Neurobiol Aging. 2015;36(2):730–9.CrossRefPubMed
55.
go back to reference Koch G, Di Lorenzo F, Bonni S, Ponzo V, Caltagirone C, Martorana A. Impaired LTP- but not LTD-like cortical plasticity in Alzheimer’s disease patients. J Alzheimers Dis. 2012;31(3):593–9.PubMed Koch G, Di Lorenzo F, Bonni S, Ponzo V, Caltagirone C, Martorana A. Impaired LTP- but not LTD-like cortical plasticity in Alzheimer’s disease patients. J Alzheimers Dis. 2012;31(3):593–9.PubMed
56.
go back to reference Bethea CL, Reddy AP. Ovarian steroids regulate gene expression related to DNA repair and neurodegenerative diseases in serotonin neurons of macaques. Mol Psychiatry. 2015;20(12):1565–78.CrossRefPubMedPubMedCentral Bethea CL, Reddy AP. Ovarian steroids regulate gene expression related to DNA repair and neurodegenerative diseases in serotonin neurons of macaques. Mol Psychiatry. 2015;20(12):1565–78.CrossRefPubMedPubMedCentral
57.
go back to reference Folch J, Patraca I, Martinez N, Pedros I, Petrov D, Ettcheto M, Abad S, Marin M, Beas-Zarate C, Camins A. The role of leptin in the sporadic form of Alzheimer’s disease. Interactions with the adipokines amylin, ghrelin and the pituitary hormone prolactin. Life Sci. 2015;140:19–28.CrossRefPubMed Folch J, Patraca I, Martinez N, Pedros I, Petrov D, Ettcheto M, Abad S, Marin M, Beas-Zarate C, Camins A. The role of leptin in the sporadic form of Alzheimer’s disease. Interactions with the adipokines amylin, ghrelin and the pituitary hormone prolactin. Life Sci. 2015;140:19–28.CrossRefPubMed
58.
go back to reference Magalhaes CA, Carvalho MG, Sousa LP, Caramelli P, Gomes KB. Leptin in Alzheimer’s disease. Clin Chim Acta. 2015;450:162–8.CrossRefPubMed Magalhaes CA, Carvalho MG, Sousa LP, Caramelli P, Gomes KB. Leptin in Alzheimer’s disease. Clin Chim Acta. 2015;450:162–8.CrossRefPubMed
59.
go back to reference de la Monte SM, Tong M. Brain metabolic dysfunction at the core of Alzheimer’s disease. Biochem Pharmacol. 2014;88(4):548–59.CrossRefPubMed de la Monte SM, Tong M. Brain metabolic dysfunction at the core of Alzheimer’s disease. Biochem Pharmacol. 2014;88(4):548–59.CrossRefPubMed
60.
go back to reference Merlo S, Spampinato S, Canonico PL, Copani A, Sortino MA. Alzheimer’s disease: brain expression of a metabolic disorder? Trends Endocrinol Metab. 2010;21(9):537–44.CrossRefPubMed Merlo S, Spampinato S, Canonico PL, Copani A, Sortino MA. Alzheimer’s disease: brain expression of a metabolic disorder? Trends Endocrinol Metab. 2010;21(9):537–44.CrossRefPubMed
61.
62.
go back to reference Perez SE, He B, Nadeem M, Wuu J, Scheff SW, Abrahamson EE, Ikonomovic MD, Mufson EJ. Resilience of precuneus neurotrophic signaling pathways despite amyloid pathology in prodromal Alzheimer’s disease. Biol Psychiatry. 2015;77(8):693–703.CrossRefPubMed Perez SE, He B, Nadeem M, Wuu J, Scheff SW, Abrahamson EE, Ikonomovic MD, Mufson EJ. Resilience of precuneus neurotrophic signaling pathways despite amyloid pathology in prodromal Alzheimer’s disease. Biol Psychiatry. 2015;77(8):693–703.CrossRefPubMed
63.
go back to reference Potter PE, Rauschkolb PK, Pandya Y, Sue LI, Sabbagh MN, Walker DG, Beach TG. Pre- and post-synaptic cortical cholinergic deficits are proportional to amyloid plaque presence and density at preclinical stages of Alzheimer’s disease. Acta Neuropathol. 2011;122(1):49–60.CrossRefPubMedPubMedCentral Potter PE, Rauschkolb PK, Pandya Y, Sue LI, Sabbagh MN, Walker DG, Beach TG. Pre- and post-synaptic cortical cholinergic deficits are proportional to amyloid plaque presence and density at preclinical stages of Alzheimer’s disease. Acta Neuropathol. 2011;122(1):49–60.CrossRefPubMedPubMedCentral
64.
go back to reference Pimenova AA, Thathiah A, De Strooper B, Tesseur I. Regulation of amyloid precursor protein processing by serotonin signaling. PLoS One. 2014;9(1):e87014.CrossRefPubMedPubMedCentral Pimenova AA, Thathiah A, De Strooper B, Tesseur I. Regulation of amyloid precursor protein processing by serotonin signaling. PLoS One. 2014;9(1):e87014.CrossRefPubMedPubMedCentral
65.
go back to reference Egorova P, Popugaeva E, Bezprozvanny I. Disturbed calcium signaling in spinocerebellar ataxias and Alzheimer’s disease. Semin Cell Dev Biol. 2015;40:127–33.CrossRefPubMedPubMedCentral Egorova P, Popugaeva E, Bezprozvanny I. Disturbed calcium signaling in spinocerebellar ataxias and Alzheimer’s disease. Semin Cell Dev Biol. 2015;40:127–33.CrossRefPubMedPubMedCentral
66.
go back to reference Scheperjans F. Can microbiota research change our understanding of neurodegenerative diseases? Neurodegener Dis Manag. 2016;6(2):81–5.CrossRefPubMed Scheperjans F. Can microbiota research change our understanding of neurodegenerative diseases? Neurodegener Dis Manag. 2016;6(2):81–5.CrossRefPubMed
67.
go back to reference Ghaisas S, Maher J, Kanthasamy A. Gut microbiome in health and disease: linking the microbiome-gut-brain axis and environmental factors in the pathogenesis of systemic and neurodegenerative diseases. Pharmacol Ther. 2016;158:52–62.CrossRefPubMed Ghaisas S, Maher J, Kanthasamy A. Gut microbiome in health and disease: linking the microbiome-gut-brain axis and environmental factors in the pathogenesis of systemic and neurodegenerative diseases. Pharmacol Ther. 2016;158:52–62.CrossRefPubMed
68.
go back to reference Catanzaro R, Anzalone M, Calabrese F, Milazzo M, Capuana M, Italia A, Occhipinti S, Marotta F. The gut microbiota and its correlations with the central nervous system disorders. Panminerva Med. 2015;57(3):127–43.PubMed Catanzaro R, Anzalone M, Calabrese F, Milazzo M, Capuana M, Italia A, Occhipinti S, Marotta F. The gut microbiota and its correlations with the central nervous system disorders. Panminerva Med. 2015;57(3):127–43.PubMed
69.
go back to reference Pedros I, Petrov D, Allgaier M, Sureda F, Barroso E, Beas-Zarate C, Auladell C, Pallas M, Vazquez-Carrera M, Casadesus G, et al. Early alterations in energy metabolism in the hippocampus of APPswe/PS1dE9 mouse model of Alzheimer’s disease. Biochim Biophys Acta. 2014;1842(9):1556–66.CrossRefPubMed Pedros I, Petrov D, Allgaier M, Sureda F, Barroso E, Beas-Zarate C, Auladell C, Pallas M, Vazquez-Carrera M, Casadesus G, et al. Early alterations in energy metabolism in the hippocampus of APPswe/PS1dE9 mouse model of Alzheimer’s disease. Biochim Biophys Acta. 2014;1842(9):1556–66.CrossRefPubMed
70.
go back to reference Zhang B, Gaiteri C, Bodea LG, Wang Z, McElwee J, Podtelezhnikov AA, Zhang C, Xie T, Tran L, Dobrin R, et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell. 2013;153(3):707–20.CrossRefPubMedPubMedCentral Zhang B, Gaiteri C, Bodea LG, Wang Z, McElwee J, Podtelezhnikov AA, Zhang C, Xie T, Tran L, Dobrin R, et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell. 2013;153(3):707–20.CrossRefPubMedPubMedCentral
71.
go back to reference Hooli BV, Kovacs-Vajna ZM, Mullin K, Blumenthal MA, Mattheisen M, Zhang C, Lange C, Mohapatra G, Bertram L, Tanzi RE. Rare autosomal copy number variations in early-onset familial Alzheimer’s disease. Mol Psychiatry. 2014;19(6):676–81.CrossRefPubMed Hooli BV, Kovacs-Vajna ZM, Mullin K, Blumenthal MA, Mattheisen M, Zhang C, Lange C, Mohapatra G, Bertram L, Tanzi RE. Rare autosomal copy number variations in early-onset familial Alzheimer’s disease. Mol Psychiatry. 2014;19(6):676–81.CrossRefPubMed
72.
go back to reference Atkin G, Hunt J, Minakawa E, Sharkey L, Tipper N, Tennant W, Paulson HL. F-box only protein 2 (Fbxo2) regulates amyloid precursor protein levels and processing. J Biol Chem. 2014;289(10):7038–48.CrossRefPubMedPubMedCentral Atkin G, Hunt J, Minakawa E, Sharkey L, Tipper N, Tennant W, Paulson HL. F-box only protein 2 (Fbxo2) regulates amyloid precursor protein levels and processing. J Biol Chem. 2014;289(10):7038–48.CrossRefPubMedPubMedCentral
73.
go back to reference Lim J, Hao T, Shaw C, Patel AJ, Szabo G, Rual JF, Fisk CJ, Li N, Smolyar A, Hill DE, et al. A protein-protein interaction network for human inherited ataxias and disorders of Purkinje cell degeneration. Cell. 2006;125(4):801–14.CrossRefPubMed Lim J, Hao T, Shaw C, Patel AJ, Szabo G, Rual JF, Fisk CJ, Li N, Smolyar A, Hill DE, et al. A protein-protein interaction network for human inherited ataxias and disorders of Purkinje cell degeneration. Cell. 2006;125(4):801–14.CrossRefPubMed
74.
go back to reference Rossini L, Hashimoto Y, Suzuki H, Kurita M, Gianfriddo M, Scali C, Roncarati R, Franceschini D, Pollio G, Trabalzini L, et al. VSTM2L is a novel secreted antagonist of the neuroprotective peptide Humanin. FASEB J. 2011;25(6):1983–2000.CrossRefPubMed Rossini L, Hashimoto Y, Suzuki H, Kurita M, Gianfriddo M, Scali C, Roncarati R, Franceschini D, Pollio G, Trabalzini L, et al. VSTM2L is a novel secreted antagonist of the neuroprotective peptide Humanin. FASEB J. 2011;25(6):1983–2000.CrossRefPubMed
75.
go back to reference Matsuoka M. Protective effects of Humanin and calmodulin-like skin protein in Alzheimer’s disease and broad range of abnormalities. Mol Neurobiol. 2015;51(3):1232–9.CrossRefPubMed Matsuoka M. Protective effects of Humanin and calmodulin-like skin protein in Alzheimer’s disease and broad range of abnormalities. Mol Neurobiol. 2015;51(3):1232–9.CrossRefPubMed
76.
go back to reference Chai GS, Duan DX, Ma RH, Shen JY, Li HL, Ma ZW, Luo Y, Wang L, Qi XH, Wang Q, et al. Humanin attenuates Alzheimer-like cognitive deficits and pathological changes induced by amyloid beta-peptide in rats. Neurosci Bull. 2014;30(6):923–35.CrossRefPubMed Chai GS, Duan DX, Ma RH, Shen JY, Li HL, Ma ZW, Luo Y, Wang L, Qi XH, Wang Q, et al. Humanin attenuates Alzheimer-like cognitive deficits and pathological changes induced by amyloid beta-peptide in rats. Neurosci Bull. 2014;30(6):923–35.CrossRefPubMed
77.
go back to reference Bertram L, McQueen MB, Mullin K, Blacker D, Tanzi RE. Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat Genet. 2007;39(1):17–23.CrossRefPubMed Bertram L, McQueen MB, Mullin K, Blacker D, Tanzi RE. Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat Genet. 2007;39(1):17–23.CrossRefPubMed
78.
go back to reference Saykin AJ, Shen L, Foroud TM, Potkin SG, Swaminathan S, Kim S, Risacher SL, Nho K, Huentelman MJ, Craig DW, et al. Alzheimer’s Disease Neuroimaging Initiative biomarkers as quantitative phenotypes: Genetics core aims, progress, and plans. Alzheimers Dement. 2010;6(3):265–73.CrossRefPubMedPubMedCentral Saykin AJ, Shen L, Foroud TM, Potkin SG, Swaminathan S, Kim S, Risacher SL, Nho K, Huentelman MJ, Craig DW, et al. Alzheimer’s Disease Neuroimaging Initiative biomarkers as quantitative phenotypes: Genetics core aims, progress, and plans. Alzheimers Dement. 2010;6(3):265–73.CrossRefPubMedPubMedCentral
79.
80.
go back to reference Bai Z, Han G, Xie B, Wang J, Song F, Peng X, Lei H. AlzBase: an integrative database for gene dysregulation in Alzheimer’s disease. Mol Neurobiol. 2016;53(1):310–9.CrossRefPubMed Bai Z, Han G, Xie B, Wang J, Song F, Peng X, Lei H. AlzBase: an integrative database for gene dysregulation in Alzheimer’s disease. Mol Neurobiol. 2016;53(1):310–9.CrossRefPubMed
81.
go back to reference Roncaglia P, Martone ME, Hill DP, Berardini TZ, Foulger RE, Imam FT, Drabkin H, Mungall CJ, Lomax J. The Gene Ontology (GO) cellular component ontology: integration with SAO (Subcellular Anatomy Ontology) and other recent developments. J Biomed Semantics. 2013;4(1):20.CrossRefPubMedPubMedCentral Roncaglia P, Martone ME, Hill DP, Berardini TZ, Foulger RE, Imam FT, Drabkin H, Mungall CJ, Lomax J. The Gene Ontology (GO) cellular component ontology: integration with SAO (Subcellular Anatomy Ontology) and other recent developments. J Biomed Semantics. 2013;4(1):20.CrossRefPubMedPubMedCentral
Metadata
Title
Analyzing the genes related to Alzheimer’s disease via a network and pathway-based approach
Authors
Yan-Shi Hu
Juncai Xin
Ying Hu
Lei Zhang
Ju Wang
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Alzheimer's Research & Therapy / Issue 1/2017
Electronic ISSN: 1758-9193
DOI
https://doi.org/10.1186/s13195-017-0252-z

Other articles of this Issue 1/2017

Alzheimer's Research & Therapy 1/2017 Go to the issue