Gene–Gene Interactions and Biological Network Analysis of Diseases with Disturbances of Human Cognitive Functions

Cover Page

Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription Access

Abstract

Neurological and mental diseases, such as schizophrenia, Alzheimer’s disease, bipolar disorder, Parkinson’s disease, have complex phenotypes with cognitive impairment. These diseases are socially significant pathologies and serious problems for world health and are distinguished by the multilevel nature of the implementation of genetic information. A number of active genes are involved in the formation of the final phenotype. Thereby, it is necessary to apply the analysis of biological networks aimed at identifying the interacting genes and proteins that lead to the pathogenesis of the disease, in order to understand the molecular mechanisms underlying the studied pathology. In this study, various online resources and databases were used to implement this approach: WebGestalt, Gene Ontology, STRING. The protein-protein interaction network was obtained, where two subnets are distinguished, one of which is involved in the risk of developing schizophrenia, and the other in the risk of developing Alzheimer’s disease.

About the authors

A. V. Bocharova

Research Institute of Medical Genetics, Tomsk National Research
Medical Center of the Russian Academy of Sciences

Author for correspondence.
Email: anna.bocharova@medgenetics.ru
Russia, 634050, Tomsk

V. A. Stepanov

Research Institute of Medical Genetics, Tomsk National Research
Medical Center of the Russian Academy of Sciences

Email: anna.bocharova@medgenetics.ru
Russia, 634050, Tomsk

References

  1. Morozova A., Zorkina Y., Abramova O. et al. Neurobiological highlights of cognitive impairment in psychiatric disorders // Int. J. Mol. Sci. 2022. V. 23. № 3. P. 1217. https://doi.org/10.3390/ijms23031217
  2. Pettersson E., Lichtenstein P., Larsson H. et al. Genetic influences on eight psychiatric disorders based on family data of 4 408 646 full and half-siblings, and genetic data of 333 748 cases and controls // Psychol. Med. 2019. V. 49. № 7. P. 1166–1173. https://doi.org/10.1017/S0033291718002039
  3. Nalls M.A., Blauwendraat C., Vallerga C.L. et al. Identification of novel risk loci, causal insights, and heritable risk for Parkinson’s disease: A meta-analysis of genome-wide association studies // Lancet Neurol. 2019. V. 18. № 12. P. 1091–1102. https://doi.org/10.1016/S1474-4422(19)30320-5
  4. Gatz M., Reynolds C.A., Fratiglioni L. et al. Role of genes and environments for explaining Alzheimer disease // Arch. Gen.Psychiatry. 2006. V. 63. № 2. P. 168–174. https://doi.org/10.1001/archpsyc.63.2.168
  5. Степанов В.А., Бочарова А.В., Марусин А.В. и др. Репликативный анализ ассоциаций генетических маркеров когнитивных признаков с болезнью Альцгеймера в российской популяции // Мол. биология. 2014. Т. 48. № 6. С. 952–962. https://doi.org/10.7868/S0026898414060160
  6. Степанов В.А., Бочарова А.В., Садуакасова К.З. и др. Репликативное исследование подверженности шизофрении с ранним началом у казахов // Генетика. 2015. Т. 51. № 2. С. 227–235. https://doi.org/10.7868/S0016675815020149
  7. Бочарова А.В., Марусин А.В., Макеева О.А. и др. Генетические варианты, связанные с нарушениями когнитивных функций человека, при болезни Альцгеймера // Мед. генетика. 2018. Т. 17. № 1. С. 14–19. https://doi.org/10.25557/2073-7998.2018.01.14-19
  8. Бочарова А.В., Марусин А.В., Иванова С.А. и др. Генетические варианты генов TCF4, LSM1 и CCDC60 ассоциированы с шизофренией // Мед. генетика. 2020. Т. 19. № 4. С. 17–19. https://doi.org/10.25557/2073-7998.2020.04.17-19
  9. Bocharova A., Vagaitseva K., Marusin F. et al. Association and gene-gene interactions study of late-onset Alzheimer’s disease in the russian population // Genes. 2021. V. 12. P. 1647. https://doi.org/10.3390/genes12101647
  10. Бочарова А.В., Степанов В.А. Генетическое разнообразие популяций Северной Евразии по маркерам, ассоциированным с заболеваниями, нарушающими когнитивные функции человека // Генетика. 2021. Т. 57. № 9. С. 1062–1072. https://doi.org/10.31857/S0016675821080026
  11. Бочарова А.В., Степанов В.А. Современные исследования генетики многофакторных заболеваний, связанных с нарушением когнитивных функций человека // Сиб. журн. клинич. и эксперим. медицины. 2021. № 4. Р. 37–44.https://doi.org/10.29001/2073-8552-2021-36-4-37-44
  12. Liao Y., Wang J., Jaehnig E.J. et al. WebGestalt 2019: Gene set analysis toolkit with revamped UIs and APIs // Nucl Acids. Res. 2019. V. 47. № W1. P. W199–W205. https://doi.org/10.1093/nar/gkz401
  13. Szklarczyk D., Gable A.L., Lyon D. et al. STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets // Nucl. Acids Res. 2019. V. 47. № D1. P. D607–D613. https://doi.org/10.1093/nar/gky1131
  14. Ashburner M., Ball C.A., Blake J.A. et al. Gene ontology: Tool for the unification of biology. The Gene Ontology Consortium // Nat. Genet. 2000. V. 25. № 1. P. 25–29. https://doi.org/10.1038/75556
  15. Bliss T.V., Cooke S.F. Long-term potentiation and long-term depression: A clinical perspective // Clinics (Sao Paulo). 2011. V. 66. Suppl. 1. P. 3–17. https://doi.org/10.1590/s1807-59322011001300002
  16. Alkadhi K.A. Neuroprotective effects of nicotine on hippocampal long-term potentiation in brain disorders // J. Pharmacol. Exp. Ther. 2018. V. 366. № 3. P. 498–508. https://doi.org/10.1124/jpet.118.247841
  17. Cleary J.P., Walsh D.M., Hofmeister J.J. et al. Natural oligomers of the amyloid-beta protein specifically disrupt cognitive function // Nat. Neurosci. 2005. V. 8. № 1. P. 79–84. https://doi.org/10.1038/nn1372
  18. Cullen W.K., Suh Y.H., Anwyl R., Rowan M.J. Block of LTP in rat hippocampus in vivo by beta-amyloid precursor protein fragments // Neuroreport. 1997. V. 8. № 15. P. 3213–3217. https://doi.org/10.1097/00001756-199710200-00006
  19. https://phgkb.cdc.gov/PHGKB/hNHome.action
  20. O’Donovan M.C., Craddock N., Norton N. et al. Identification of loci associated with schizophrenia by genome-wide association and follow-up // Nat. Genet. 2008. V. 40. № 9. P. 1053–1055. https://doi.org/10.1038/ng.201
  21. Matthews J.M., Sunde M. Zinc fingers-folds for many occasions // IUBMB Life. 2002. V. 54. № 6. P. 351–355. https://doi.org/10.1080/15216540216035
  22. Федотова А.А., Бончук А.Н., Могила В.A., Георгиев П.Г. Белки с цинковыми пальцами типа С2Н2 – самый многочисленный и наименее изученный класс транскрипционных факторов высших эукариот // Acta Naturae. 2017. Т. 9. № 2. С. 50–61.
  23. Deans P.J.M., Raval P., Sellers K.J. et al. Psychosis risk candidate ZNF804A localizes to synapses and regulates neurite formation and dendritic spine structure // Biol. Psychiatry. 2017. V. 82. № 1. P. 49–61. https://doi.org/10.1016/j.biopsych.2016.08.038
  24. Penzes P., Cahill M.E., Jones K.A. et al. Dendritic spine pathology in neuropsychiatric disorders // Nat. Neurosci. 2011. V. 14. № 3. P. 285–293. https://doi.org/10.1038/nn.2741
  25. Chang H., Xiao X., Li M. The schizophrenia risk gene ZNF804A: Clinical associations, biological mechanisms and neuronal functions // Mol. Psychiatry. 2017. V. 22. № 7. P. 944–953. https://doi.org/10.1038/mp.2017.19
  26. Srivastava D.P., Woolfrey K.M., Penzes P. Analysis of dendritic spine morphology in cultured CNS neurons // J. Vis. Exp. 2011. V. 53. P. e2794. https://doi.org/10.3791/2794
  27. Zhou D., Xiao X., Li M. The schizophrenia risk isoform ZNF804AE3E4 affects dendritic spine // Schizophrenia Res. 2020. V. 218. P. 324–325. https://doi.org/10.1016/j.schres.2019.12.038
  28. Tao R., Cousijn H., Jaffe A.E. et al. Expression of ZNF804A in human brain and alterations in schizophrenia, bipolar disorder, and major depressive disorder: A novel transcript fetally regulated by the psychosis risk variant rs1344706 // JAMA Psychiat. 2014. V. 71. № 10. P. 1112–1120. https://doi.org/10.1001/jamapsychiatry.2014.1079
  29. Corder E.H., Saunders A.M., Strittmatter W.J. et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families // Science. 1993. V. 261. № 5123. P. 921–923. https://doi.org/10.1126/science.8346443
  30. Strittmatter W.J., Weisgraber K.H., Huang D.Y. et al. Binding of human apolipoprotein E to synthetic amyloid beta peptide: Isoform-specific effects and implications for late-onset Alzheimer disease // Proc. Natl Acad. Sci. USA. 1993. V. 90. № 17. P. 8098–8102. https://doi.org/10.1073/pnas.90.17.8098
  31. Roses A.D. Apolipoprotein E alleles as risk factors in Alzheimer’s disease // Annu. Rev. Med. 1996. V. 47. P. 387–400. https://doi.org/10.1146/annurev.med.47.1.387
  32. Kecmanović M., Dobricić V., Dimitrijević R. et al. Schizophrenia and apolipoprotein E gene polymorphism in Serbian population // Int. J.Neurosci. 2010. V. 120. № 7. P. 502–506. https://doi.org/10.3109/00207451003765956
  33. Gibbons A.S., Udawela M., Jeon W.J. et al. The neurobiology of APOE in schizophrenia and mood disorders // Front Biosci. (Landmark Ed). 2011. V. 16. P. 962–979. https://doi.org/10.2741/3729
  34. Giau V.V., Bagyinszky E., An S.S., Kim S.Y. Role of apolipoprotein E in neurodegenerative diseases // Neuropsychiatr. Dis. Treat. 2015. V. 11. P. 1723–1737. https://doi.org/10.2147/NDT.S84266
  35. Yin Y., Wang Z. ApoE and neurodegenerative diseases in aging // Adv. Exp. Med. Biol. 2018. V. 1086. P. 77–92. https://doi.org/10.1007/978-981-13-1117-8_5
  36. Xu M.Q., St Clair D., He L. Meta-analysis of association between ApoE epsilon4 allele and schizophrenia // Schizophr. Res. 2006. V. 84. № 2–3. P. 228–235. https://doi.org/10.1016/j.schres.2006.02.015
  37. Allen N.C., Bagade S., McQueen M.B. et al. Systematic meta-analyses and field synopsis of genetic association studies in schizophrenia: the SzGene database // Nat. Genet. 2008. V. 40. № 7. P. 827–834. https://doi.org/10.1038/ng.171
  38. Lambert J.C., Ibrahim-Verbaas C.A., Harold D. et al. Meta-analysis of 74 046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease // Nat. Genet. 2013. V. 45. № 12. P. 1452–1458. https://doi.org/10.1038/ng.2802
  39. Abyadeh M., Djafarian K., Heydarinejad F. et al. Association between apolipoprotein E gene polymorphism and Alzheimer’s disease in an Iranian population: A Meta-Analysis // J. Mol. Neurosci. 2019. V. 69. № 4. P. 557–562. https://doi.org/10.1007/s12031-019-01381-1
  40. Ishii K., Kubo K., Nakajima K. Reelin and neuropsychiatric disorders // Front. Cell Neurosci. 2016. V. 10. P. 229. https://doi.org/10.3389/fncel.2016.00229
  41. Ovadia G., Shifman S. The genetic variation of RELN expression in schizophrenia and bipolar disorder // PLoS One. 2011. V. 6. P. e19955. https://doi.org/10.1371/journal.pone.0019955
  42. Bufill E., Roura-Poch P., Sala-Matavera I. et al. Reelin signaling pathway genotypes and Alzheimer disease in a Spanish population // Alzheimer Dis. Assoc. Disord. 2015. V. 29. № 2. P. 169–172. https://doi.org/10.1097/WAD.0000000000000002
  43. Saez-Valero J., Costell M., Sjogren M. et al. Altered levels of cerebrospinal fluid reelin in frontotemporal dementia and Alzheimer’s disease// J. Neurosci. Res. 2003. V. 72. № 1. P. 132–136. https://doi.org/10.1002/jnr.10554
  44. Seripa D., Matera M.G., Franceschi M. et al. The RELN locus in Alzheimer’s disease // J.Alzheimers. Dis. 2008. V. 14. № 3. P. 335–344. https://doi.org/10.3233/jad-2008-14308
  45. Botella-López A., Cuchillo-Ibáñez I., Cotrufo T. et al. Beta-amyloid controls altered Reelin expression and processing in Alzheimer’s disease // Neurobiol. Dis. 2010. V. 37. № 3. P. 682–691. https://doi.org/10.1016/j.nbd.2009.12.006
  46. Pujadas L., Rossi D., Andrés R. et al. Reelin delays amyloid-beta fibril formation and rescues cognitive deficits in a model of Alzheimer’s disease // Nat. Commun. 2014. V. 5. P. 3443. https://doi.org/10.1038/ncomms4443
  47. Rossi D., Gruart A., Contreras-Murillo G. et al. Reelin reverts biochemical, physiological and cognitive alterations in mouse models of Tauopathy // Prog. Neurobiol. 2020. V. 186. P. 101743. https://doi.org/10.1016/j.pneurobio.2019.101743
  48. Roth T.L., Lubin F.D., Sodhi M., Kleinman J.E. Epigenetic mechanisms in schizophrenia // Biochim. Biophys. Acta. 2009. V. 1790. P. 869–877. https://doi.org/10.1016/j.bbagen.2009.06.009
  49. Veldic M., Guidotti A., Maloku E. et al. In psychosis, cortical interneurons overexpress DNA-methyltransferase 1 // Proc. Natl Acad. Sci. USA. 2005. V. 102. № 6. P. 2152–2157. https://doi.org/10.1073/pnas.0409665102
  50. Impagnatiello F., Caruncho H., Niu S. et al. Reelin promotes hippocampal dendrite development through the VLDLR/ApoER2-Dab1 pathway // Neuron. 2004. V. 41. P. 71–84. https://doi.org/10.1016/s0896-6273(03)00819-5
  51. Glantz L.A., Lewis D.A. Decreased dendritic spine density on prefrontal cortical pyramidal neurons in schizophrenia // Arch. Gen. Psychiatry. 2000. V. 57. P. 65–73. https://doi.org/10.1001/archpsyc.57.1.65

Supplementary files

Supplementary Files
Action
1. JATS XML
2.

Download (488KB)

Copyright (c) 2023 А.В. Бочарова, В.А. Степанов

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies