#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Accelerated brain aging towards transcriptional inversion in a zebrafish model of the K115fs mutation of human PSEN2


Autoři: Nhi Hin aff001;  Morgan Newman aff002;  Jan Kaslin aff003;  Alon M. Douek aff003;  Amanda Lumsden aff004;  Seyed Hani Moussavi Nik aff001;  Yang Dong aff001;  Xin-Fu Zhou aff005;  Noralyn B. Mañucat-Tan aff005;  Alastair Ludington aff001;  David L. Adelson aff006;  Stephen Pederson aff001;  Michael Lardelli aff002
Působiště autorů: Bioinformatics Hub, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia aff001;  Alzheimer’s Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia aff002;  Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia aff003;  College of Medicine and Public Health, and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia aff004;  School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia aff005;  Centre for Bioinformatics and Computational Genetics, School of Bioogical Sciences, Adelaide, South Australia, Australia aff006
Vyšlo v časopise: PLoS ONE 15(1)
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.pone.0227258

Souhrn

Background

The molecular changes involved in Alzheimer’s disease (AD) progression remain unclear since we cannot easily access antemortem human brains. Some non-mammalian vertebrates such as the zebrafish preserve AD-relevant transcript isoforms of the PRESENILIN genes lost from mice and rats. One example is PS2V, the alternative transcript isoform of the PSEN2 gene. PS2V is induced by hypoxia/oxidative stress and shows increased expression in late onset, sporadic AD brains. A unique, early onset familial AD mutation of PSEN2, K115fs, mimics the PS2V coding sequence suggesting that forced, early expression of PS2V-like isoforms may contribute to AD pathogenesis. Here we use zebrafish to model the K115fs mutation to investigate the effects of forced PS2V-like expression on the transcriptomes of young adult and aged adult brains.

Methods

We edited the zebrafish genome to model the K115fs mutation. To explore its effects at the molecular level, we analysed the brain transcriptome and proteome of young (6-month-old) and aged (24-month-old) wild type and heterozygous mutant female sibling zebrafish. Finally, we used gene co-expression network analysis (WGCNA) to compare molecular changes in the brains of these fish to human AD.

Results

Young heterozygous mutant fish show transcriptional changes suggesting accelerated brain aging and increased glucocorticoid signalling. These early changes precede a transcriptional ‘inversion’ that leads to glucocorticoid resistance and other likely pathological changes in aged heterozygous mutant fish. Notably, microglia-associated immune responses regulated by the ETS transcription factor family are altered in both our zebrafish mutant model and in human AD. The molecular changes we observe in aged heterozygous mutant fish occur without obvious histopathology and possibly in the absence of Aβ.

Conclusions

Our results suggest that forced expression of a PS2V-like isoform contributes to immune and stress responses favouring AD pathogenesis. This highlights the value of our zebrafish genetic model for exploring molecular mechanisms involved in AD pathogenesis.

Klíčová slova:

Alzheimer's disease – Gene expression – Genetic networks – Immune response – Mutation – Neural networks – Polymerase chain reaction – Zebrafish


Zdroje

1. Cadonic C, Sabbir MG, Albensi BC. Mechanisms of Mitochondrial Dysfunction in Alzheimer’s Disease. Mol Neurobiol. 2015. doi: 10.1007/s12035-015-9515-5 26537901.

2. Castellani R, Hirai K, Aliev G, Drew KL, Nunomura A, Takeda A, et al. Role of mitochondrial dysfunction in Alzheimer’s disease. J Neurosci Res. 2002;70(3):357–60. Epub 2002/10/23. doi: 10.1002/jnr.10389 12391597.

3. Fedrizzi L, Carafoli E. Ca2+ dysfunction in neurodegenerative disorders: Alzheimer’s disease. Biofactors. 2011;37(3):189–96. Epub 2011/06/24. doi: 10.1002/biof.157 21698698.

4. Mills E, Dong XP, Wang F, Xu H. Mechanisms of brain iron transport: insight into neurodegeneration and CNS disorders. Future Med Chem. 2010;2(1):51–64. doi: 10.4155/fmc.09.140 20161623.

5. Moir RD, Atwood CS, Huang X, Tanzi RE, Bush AI. Mounting evidence for the involvement of zinc and copper in Alzheimer’s disease. Eur J Clin Invest. 1999;29(7):569–70. Epub 1999/07/20. doi: 10.1046/j.1365-2362.1999.00472.x 10411660.

6. Arimon M, Takeda S, Post KL, Svirsky S, Hyman BT, Berezovska O. Oxidative stress and lipid peroxidation are upstream of amyloid pathology. Neurobiol Dis. 2015;84:109–19. doi: 10.1016/j.nbd.2015.06.013 26102023.

7. Oresic M, Hyotylainen T, Herukka SK, Sysi-Aho M, Mattila I, Seppanan-Laakso T, et al. Metabolome in progression to Alzheimer’s disease. Transl Psychiatry. 2011;1:e57. Epub 2011/01/01. doi: 10.1038/tp.2011.55 22832349.

8. Poirier J, Miron J, Picard C, Gormley P, Theroux L, Breitner J, et al. Apolipoprotein E and lipid homeostasis in the etiology and treatment of sporadic Alzheimer’s disease. Neurobiol Aging. 2014;35 Suppl 2:S3–10. Epub 2014/06/29. doi: 10.1016/j.neurobiolaging.2014.03.037 24973118.

9. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14(4):388–405. Epub 2015/03/21. doi: 10.1016/S1474-4422(15)70016-5 25792098.

10. Zotova E, Nicoll JA, Kalaria R, Holmes C, Boche D. Inflammation in Alzheimer’s disease: relevance to pathogenesis and therapy. Alzheimers Res Ther. 2010;2(1):1. Epub 2010/02/04. doi: 10.1186/alzrt24 20122289.

11. Abramsson A, Kettunen P, Banote RK, Lott E, Li M, Arner A, et al. The zebrafish amyloid precursor protein-b is required for motor neuron guidance and synapse formation. Dev Biol. 2013;381(2):377–88. doi: 10.1016/j.ydbio.2013.06.026 23850871.

12. De Strooper B. Proteases and proteolysis in Alzheimer disease: a multifactorial view on the disease process. Physiol Rev. 2010;90(2):465–94. doi: 10.1152/physrev.00023.2009 20393191.

13. Hipp MS, Park SH, Hartl FU. Proteostasis impairment in protein-misfolding and -aggregation diseases. Trends Cell Biol. 2014. Epub 2014/06/21. doi: 10.1016/j.tcb.2014.05.003 24946960.

14. Braak H, Braak E. Diagnostic criteria for neuropathologic assessment of Alzheimer’s disease. Neurobiol Aging. 1997;18(4 Suppl):S85–8. Epub 1997/07/01. doi: 10.1016/s0197-4580(97)00062-6 9330992.

15. Masters CL, Bateman RJ, Blennow K, Rowe CC, Sperling RA, Cummings JL. Alzheimer’s disease. Nature reviews Disease Primers. 2015;1:1–8. doi: 10.1038/nrdp.2015.56 27188934

16. Mehta D, Jackson R, Paul G, Shi J, Sabbagh M. Why do trials for Alzheimer’s disease drugs keep failing? A discontinued drug perspective for 2010–2015. Expert Opin Investig Drugs. 2017;26(6):735–9. doi: 10.1080/13543784.2017.1323868 28460541.

17. Villemagne VL, Burnham S, Bourgeat P, Brown B, Ellis KA, Salvado O, et al. Amyloid beta deposition, neurodegeneration, and cognitive decline in sporadic Alzheimer’s disease: a prospective cohort study. Lancet Neurol. 2013;12(4):357–67. Epub 2013/03/13. doi: 10.1016/S1474-4422(13)70044-9 23477989.

18. Bateman RJ, Xiong C, Benzinger TL, Fagan AM, Goate A, Fox NC, et al. Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. N Engl J Med. 2012;367(9):795–804. doi: 10.1056/NEJMoa1202753 22784036.

19. Iturria-Medina Y, Sotero RC, Toussaint PJ, Mateos-Perez JM, Evans AC, Alzheimer’s Disease Neuroimaging I. Early role of vascular dysregulation on late-onset Alzheimer’s disease based on multifactorial data-driven analysis. Nat Commun. 2016;7:11934. doi: 10.1038/ncomms11934 27327500.

20. Quiroz YT, Schultz AP, Chen K, Protas HD, Brickhouse M, Fleisher AS, et al. Brain Imaging and Blood Biomarker Abnormalities in Children With Autosomal Dominant Alzheimer Disease: A Cross-Sectional Study. JAMA Neurol. 2015;72(8):912–9. doi: 10.1001/jamaneurol.2015.1099 26121081.

21. Reiman EM, Quiroz YT, Fleisher AS, Chen K, Velez-Pardo C, Jimenez-Del-Rio M, et al. Brain imaging and fluid biomarker analysis in young adults at genetic risk for autosomal dominant Alzheimer’s disease in the presenilin 1 E280A kindred: a case-control study. Lancet Neurol. 2012;11(12):1048–56. doi: 10.1016/S1474-4422(12)70228-4 23137948.

22. Reiman EM, Chen K, Alexander GE, Caselli RJ, Bandy D, Osborne D, et al. Functional brain abnormalities in young adults at genetic risk for late-onset Alzheimer’s dementia. Proc Natl Acad Sci U S A. 2004;101(1):284–9. Epub 2003/12/23. doi: 10.1073/pnas.2635903100 14688411

23. Berchtold NC, Sabbagh MN, Beach TG, Kim RC, Cribbs DH, Cotman CW. Brain gene expression patterns differentiate mild cognitive impairment from normal aged and Alzheimer’s disease. Neurobiol Aging. 2014;35(9):1961–72. doi: 10.1016/j.neurobiolaging.2014.03.031 24786631.

24. Antonell A, Llado A, Altirriba J, Botta-Orfila T, Balasa M, Fernandez M, et al. A preliminary study of the whole-genome expression profile of sporadic and monogenic early-onset Alzheimer’s disease. Neurobiol Aging. 2013;34(7):1772–8. Epub 2013/02/02. doi: 10.1016/j.neurobiolaging.2012.12.026 23369545.

25. Guo Q, Fu W, Sopher BL, Miller MW, Ware CB, Martin GM, et al. Increased vulnerability of hippocampal neurons to excitotoxic necrosis in presenilin-1 mutant knock-in mice. Nat Med. 1999;5(1):101–6. doi: 10.1038/4789 9883847.

26. Kawasumi M, Chiba T, Yamada M, Miyamae-Kaneko M, Matsuoka M, Nakahara J, et al. Targeted introduction of V642I mutation in amyloid precursor protein gene causes functional abnormality resembling early stage of Alzheimer’s disease in aged mice. Eur J Neurosci. 2004;19(10):2826–38. doi: 10.1111/j.0953-816X.2004.03397.x 15147316.

27. Siman R, Reaume AG, Savage MJ, Trusko S, Lin YG, Scott RW, et al. Presenilin-1 P264L knock-in mutation: differential effects on abeta production, amyloid deposition, and neuronal vulnerability. J Neurosci. 2000;20(23):8717–26. doi: 10.1523/JNEUROSCI.20-23-08717.2000 11102478.

28. Hargis KE, Blalock EM. Transcriptional signatures of brain aging and Alzheimer’s disease: What are our rodent models telling us? Behav Brain Res. 2017;322(Pt B):311–28. Epub 2016/05/04. doi: 10.1016/j.bbr.2016.05.007 27155503.

29. Sato N, Hori O, Yamaguchi A, Lambert JC, Chartier-Harlin MC, Robinson PA, et al. A novel presenilin-2 splice variant in human Alzheimer’s disease brain tissue. Journal of Neurochemistry. 1999;72(6):2498–505. doi: 10.1046/j.1471-4159.1999.0722498.x 10349860

30. Jayadev S, Leverenz JB, Steinbart E, Stahl J, Klunk W, Yu CE, et al. Alzheimer’s disease phenotypes and genotypes associated with mutations in presenilin 2. Brain. 2010;133:1143–54. doi: 10.1093/brain/awq033 20375137

31. Newman M, Wilson L, Verdile G, Lim A, Khan I, Moussavi Nik SH, et al. Differential, dominant activation and inhibition of Notch signalling and APP cleavage by truncations of PSEN1 in human disease. Hum Mol Genet. 2014;23(3):602–17. Epub 2013/10/09. doi: 10.1093/hmg/ddt448 24101600.

32. Moussavi Nik SH, Newman M, Wilson L, Ebrahimie E, Wells S, Musgrave I, et al. Alzheimer’s disease-related peptide PS2V plays ancient, conserved roles in suppression of the unfolded protein response under hypoxia and stimulation of gamma-secretase activity. Hum Mol Genet. 2015. Epub 2015/03/31. doi: 10.1093/hmg/ddv110 25814654.

33. Sharman MJ, Moussavi Nik SH, Chen MM, Ong D, Wijaya L, Laws SM, et al. The Guinea Pig as a Model for Sporadic Alzheimer’s Disease (AD): The Impact of Cholesterol Intake on Expression of AD-Related Genes. PLoS One. 2013;8(6):e66235. doi: 10.1371/journal.pone.0066235 23805206.

34. Seshadri S, Wolf PA, Beiser A, Au R, McNulty K, White R, et al. Lifetime risk of dementia and Alzheimer’s disease. The impact of mortality on risk estimates in the Framingham Study. Neurology. 1997;49(6):1498–504. doi: 10.1212/wnl.49.6.1498 9409336.

35. Gamberger D, Ženko B, Mitelpunkt A, Shachar N, Lavrač N. Clusters of male and female Alzheimer’s disease patients in the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database. Brain Inform. 2016;3(3):169–79. Epub 2016/03/30. doi: 10.1007/s40708-016-0035-5 27525218.

36. Oosterhof N, Holtman IR, Kuil LE, van der Linde HC, Boddeke EW, Eggen BJ, et al. Identification of a conserved and acute neurodegeneration-specific microglial transcriptome in the zebrafish. Glia. 2017;65(1):138–49. Epub 2016/10/19. doi: 10.1002/glia.23083 27757989.

37. Lein ES, Hawrylycz MJ, Ao N, Ayres M, Bensinger A, Bernard A, et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature. 2007;445(7124):168–76. Epub 2006/12/06. doi: 10.1038/nature05453 17151600.

38. Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1(6):417–25. doi: 10.1016/j.cels.2015.12.004 26771021.

39. de Sousa Abreu R, Penalva LO, Marcotte EM, Vogel C. Global signatures of protein and mRNA expression levels. Mol Biosyst. 2009;5(12):1512–26. Epub 2009/10/01. doi: 10.1039/b908315d 20023718.

40. Kumar D, Bansal G, Narang A, Basak T, Abbas T, Dash D. Integrating transcriptome and proteome profiling: Strategies and applications. Proteomics. 2016;16(19):2533–44. Epub 2016/08/25. doi: 10.1002/pmic.201600140 27343053.

41. Allen M, Carrasquillo MM, Funk C, Heavner BD, Zou F, Younkin CS, et al. Human whole genome genotype and transcriptome data for Alzheimer’s and other neurodegenerative diseases. Sci Data. 2016;3:160089. Epub 2016/10/11. doi: 10.1038/sdata.2016.89 27727239.

42. Ballouz S, Verleyen W, Gillis J. Guidance for RNA-seq co-expression network construction and analysis: safety in numbers. Bioinformatics. 2015;31(13):2123–30. doi: 10.1093/bioinformatics/btv118 25717192.

43. Zhang B, Horvath S. A General Framework for Weighted Gene Co-Expression Network Analysis. Statistical Applications in Genetics and Molecular Biology2005.

44. Hawrylycz MJ, Lein ES, Guillozet-Bongaarts AL, Shen EH, Ng L, Miller JA, et al. An anatomically comprehensive atlas of the adult human brain transcriptome. Nature. 2012;489(7416):391–9. doi: 10.1038/nature11405 22996553.

45. Winden KD, Oldham MC, Mirnics K, Ebert PJ, Swan CH, Levitt P, et al. The organization of the transcriptional network in specific neuronal classes. Mol Syst Biol. 2009;5:291. doi: 10.1038/msb.2009.46 19638972.

46. Oldham MC, Horvath S, Geschwind DH. Conservation and evolution of gene coexpression networks in human and chimpanzee brains. Proceedings of the National Academy of Sciences. 2006;103(47):17973–8. doi: 10.1073/pnas.0605938103 17101986

47. Miller JA, Horvath S, Geschwind DH. Divergence of human and mouse brain transcriptome highlights Alzheimer disease pathways. Proceedings of the National Academy of Sciences. 2010;107(28):12698–703.

48. Zhang B, Gaiteri C, Bodea LG, Wang Z, McElwee J, Podtelezhnikov AA, et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell. 2013;153(3):707–20. doi: 10.1016/j.cell.2013.03.030 23622250.

49. Langfelder P, Luo R, Oldham MC, Horvath S. Is my network module preserved and reproducible? PLoS Comput Biol. 2011;7(1):e1001057. doi: 10.1371/journal.pcbi.1001057 21283776.

50. Solchenberger B, Russell C, Kremmer E, Haass C, Schmid B. Granulin knock out zebrafish lack frontotemporal lobar degeneration and neuronal ceroid lipofuscinosis pathology. PLoS One. 2015;10(3):e0118956. doi: 10.1371/journal.pone.0118956 25785851.

51. Moore DB, Gillentine MA, Botezatu NM, Wilson KA, Benson AE, Langeland JA. Asynchronous evolutionary origins of Abeta and BACE1. Mol Biol Evol. 2014;31(3):696–702. doi: 10.1093/molbev/mst262 24361992.

52. Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;256(5054):184–5. doi: 10.1126/science.1566067 1566067.

53. Blalock EM, Geddes JW, Chen KC, Porter NM, Markesbery WR, Landfield PW. Incipient Alzheimer’s disease: microarray correlation analyses reveal major transcriptional and tumor suppressor responses. Proc Natl Acad Sci U S A. 2004;101(7):2173–8. doi: 10.1073/pnas.0308512100 14769913.

54. Sperling RA, Dickerson BC, Pihlajamaki M, Vannini P, LaViolette PS, Vitolo OV, et al. Functional alterations in memory networks in early Alzheimer’s disease. Neuromolecular Med. 2010;12(1):27–43. doi: 10.1007/s12017-009-8109-7 20069392.

55. Head E, Lott IT, Patterson D, Doran E, Haier RJ. Possible compensatory events in adult Down syndrome brain prior to the development of Alzheimer disease neuropathology: targets for nonpharmacological intervention. J Alzheimers Dis. 2007;11(1):61–76. doi: 10.3233/jad-2007-11110 17361036.

56. Du X, Pang TY. Is Dysregulation of the HPA-Axis a Core Pathophysiology Mediating Co-Morbid Depression in Neurodegenerative Diseases? Front Psychiatry. 2015;6:32. doi: 10.3389/fpsyt.2015.00032 25806005.

57. Silverman MN, Sternberg EM. Glucocorticoid regulation of inflammation and its functional correlates: from HPA axis to glucocorticoid receptor dysfunction. Ann N Y Acad Sci. 2012;1261:55–63. doi: 10.1111/j.1749-6632.2012.06633.x 22823394.

58. Arlt S, Demiralay C, Tharun B, Geisel O, Storm N, Eichenlaub M, et al. Genetic risk factors for depression in Alzheimer`s disease patients. Curr Alzheimer Res. 2013;10(1):72–81. 23157339.

59. Lin JX, Leonard WJ. The role of Stat5a and Stat5b in signaling by IL-2 family cytokines. Oncogene. 2000;19(21):2566–76. doi: 10.1038/sj.onc.1203523 10851055.

60. Moon RT, Bowerman B, Boutros M, Perrimon N. The promise and perils of Wnt signaling through beta-catenin. Science. 2002;296(5573):1644–6. doi: 10.1126/science.1071549 12040179

61. Rapoport SI, Hatanpaa K, Brady DR, Chandrasekaran K. Brain energy metabolism, cognitive function and down-regulated oxidative phosphorylation in Alzheimer disease. Neurodegeneration. 1996;5(4):473–6. doi: 10.1006/neur.1996.0065 9117565.

62. Barron AM, Fuller SJ, Verdile G, Martins RN. Reproductive hormones modulate oxidative stress in Alzheimer’s disease. Antioxid Redox Signal. 2006;8(11–12):2047–59. doi: 10.1089/ars.2006.8.2047 17034349.

63. Schapira AH. Oxidative stress and mitochondrial dysfunction in neurodegeneration. Curr Opin Neurol. 1996;9(4):260–4. doi: 10.1097/00019052-199608000-00003 8858182.

64. Perry G, Nunomura A, Hirai K, Takeda A, Aliev G, Smith MA. Oxidative damage in Alzheimer’s disease: the metabolic dimension. Int J Dev Neurosci. 2000;18(4–5):417–21. Epub 2000/05/19. doi: 10.1016/s0736-5748(00)00006-x 10817925

65. Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, et al. Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol. 2001;60(8):759–67. Epub 2001/08/07. doi: 10.1093/jnen/60.8.759 11487050.

66. Perry G, Nunomura A, Hirai K, Zhu X, Perez M, Avila J, et al. Is oxidative damage the fundamental pathogenic mechanism of Alzheimer’s and other neurodegenerative diseases? Free Radic Biol Med. 2002;33(11):1475–9. Epub 2002/11/26. doi: 10.1016/s0891-5849(02)01113-9 12446204

67. Scherz-Shouval R, Elazar Z. ROS, mitochondria and the regulation of autophagy. Trends Cell Biol. 2007;17(9):422–7. Epub 2007/09/07. doi: 10.1016/j.tcb.2007.07.009 17804237.

68. Daulatzai MA. Death by a thousand cuts in Alzheimer’s disease: hypoxia-the prodrome. Neurotox Res. 2013;24(2):216–43. doi: 10.1007/s12640-013-9379-2 23400634.

69. Daulatzai MA. Cerebral hypoperfusion and glucose hypometabolism: Key pathophysiological modulators promote neurodegeneration, cognitive impairment, and Alzheimer’s disease. J Neurosci Res. 2017;95(4):943–72. doi: 10.1002/jnr.23777 27350397.

70. Zhao WQ, Ravindranath L, Mohamed AS, Zohar O, Chen GH, Lyketsos CG, et al. MAP kinase signaling cascade dysfunction specific to Alzheimer’s disease in fibroblasts. Neurobiol Dis. 2002;11(1):166–83. doi: 10.1006/nbdi.2002.0520 12460556.

71. Drewes G, Lichtenberg-Kraag B, Döring F, Mandelkow EM, Biernat J, Goris J, et al. Mitogen activated protein (MAP) kinase transforms tau protein into an Alzheimer-like state. EMBO J. 1992;11(6):2131–8. 1376245.

72. Maroulakou IG, Bowe DB. Expression and function of Ets transcription factors in mammalian development: a regulatory network. Oncogene. 2000;19(55):6432–42. doi: 10.1038/sj.onc.1204039 11175359.

73. Kar A, Gutierrez-Hartmann A. Molecular mechanisms of ETS transcription factor-mediated tumorigenesis. Crit Rev Biochem Mol Biol. 2013;48(6):522–43. doi: 10.3109/10409238.2013.838202 24066765.

74. Gjoneska E, Pfenning AR, Mathys H, Quon G, Kundaje A, Tsai LH, et al. Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer’s disease. Nature. 2015;518(7539):365–9. doi: 10.1038/nature14252 25693568.

75. Morgan SC, Taylor DL, Pocock JM. Microglia release activators of neuronal proliferation mediated by activation of mitogen-activated protein kinase, phosphatidylinositol-3-kinase/Akt and delta-Notch signalling cascades. J Neurochem. 2004;90(1):89–101. doi: 10.1111/j.1471-4159.2004.02461.x 15198670.

76. Herculano-Houzel S. Scaling of brain metabolism with a fixed energy budget per neuron: implications for neuronal activity, plasticity and evolution. PLoS One. 2011;6(3):e17514. doi: 10.1371/journal.pone.0017514 21390261.

77. Yamada T, Sasaki H, Furuya H, Miyata T, Goto I, Sakaki Y. Complementary DNA for the mouse homolog of the human amyloid beta protein precursor. Biochem Biophys Res Commun. 1987;149(2):665–71. doi: 10.1016/0006-291x(87)90419-0 3322280.

78. Musa A, Lehrach H, Russo VA. Distinct expression patterns of two zebrafish homologues of the human APP gene during embryonic development. Dev Genes Evol. 2001;211(11):563–7. Epub 2002/02/28. doi: 10.1007/s00427-001-0189-9 11862463.

79. Monsell SE, Kukull WA, Roher AE, Maarouf CL, Serrano G, Beach TG, et al. Characterizing Apolipoprotein E epsilon4 Carriers and Noncarriers With the Clinical Diagnosis of Mild to Moderate Alzheimer Dementia and Minimal beta-Amyloid Peptide Plaques. JAMA Neurol. 2015;72(10):1124–31. doi: 10.1001/jamaneurol.2015.1721 26302353.

80. Jack CR Jr., Albert MS, Knopman DS, McKhann GM, Sperling RA, Carrillo MC, et al. Introduction to the recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):257–62. 21514247.

81. Whitehouse PJ, George DR. A tale of two reports: what recent publications from the Alzheimer’s Association and Institute of Medicine say about the state of the field. J Alzheimers Dis. 2016;49(1):21–5. doi: 10.3233/JAD-150663 26444797.

82. Morris GP, Clark IA, Vissel B. Questions concerning the role of amyloid-β in the definition, aetiology and diagnosis of Alzheimer’s disease. Acta Neuropathol. 2018;136(5):663–89. Epub 2018/10/22. doi: 10.1007/s00401-018-1918-8 30349969.

83. Jansen WJ, Ossenkoppele R, Knol DL, Tijms BM, Scheltens P, Verhey FR, et al. Prevalence of cerebral amyloid pathology in persons without dementia: a meta-analysis. JAMA. 2015;313(19):1924–38. doi: 10.1001/jama.2015.4668 25988462.

84. Braggin JE, Bucks SA, Course MM, Smith CL, Sopher B, Osnis L, et al. Alternative splicing in a presenilin 2 variant associated with Alzheimer disease. Ann Clin Transl Neurol. 2019;6(4):762–77. Epub 2019/03/10. doi: 10.1002/acn3.755 31020001.

85. Nornes S, Newman M, Verdile G, Wells S, Stoick-Cooper CL, Tucker B, et al. Interference with splicing of Presenilin transcripts has potent dominant negative effects on Presenilin activity. Hum Mol Genet. 2008;17(3):402–12. Epub 2007/11/06. doi: 10.1093/hmg/ddm317 17981814.

86. Newman M, Hin N, Pederson S, Lardelli M. Brain transcriptome analysis of a familial Alzheimer’s disease-like mutation in the zebrafish presenilin 1 gene implies effects on energy production. Mol Brain. 2019;12(1):43. Epub 2019/05/03. doi: 10.1186/s13041-019-0467-y 31053140.

87. Huggett JF, Whale A. Digital PCR as a novel technology and its potential implications for molecular diagnostics. Clin Chem. 2013;59(12):1691–3. Epub 2013/10/07. doi: 10.1373/clinchem.2013.214742 24100808.

88. de St Groth Fazekas. The evaluation of limiting dilution assays. J Immunol Methods. 1982;49(2):R11–23. doi: 10.1016/0022-1759(82)90269-1 7040548.

89. Wisniewski JR, Zougman A, Nagaraj N, Mann M. Universal sample preparation method for proteome analysis. Nat Methods. 2009;6(5):359–62. doi: 10.1038/nmeth.1322 19377485.

90. Cox J, Hein MY, Luber CA, Paron I, Nagaraj N, Mann M. Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ. Mol Cell Proteomics. 2014;13(9):2513–26. doi: 10.1074/mcp.M113.031591 24942700.

91. Andrews S. FastQC. 0.11.5 ed2010.

92. Lindgreen S. AdapterRemoval: easy cleaning of next-generation sequencing reads. BMC Res Notes. 2012;5:337. doi: 10.1186/1756-0500-5-337 22748135.

93. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015;12(4):357–60. doi: 10.1038/nmeth.3317 25751142.

94. Broad Institute. Picard. 2.14.0 ed2017. p. A set of command line tools (in Java) for manipulating high-throughput sequencing (HTS) data and formats such as SAM/BAM/CRAM and VCF.

95. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30(7):923–30. doi: 10.1093/bioinformatics/btt656 24227677.

96. R Core Team. R: A Language and Environment for Statistical Computing. R Foundation for Statistical Computing; 2017.

97. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40. doi: 10.1093/bioinformatics/btp616 19910308.

98. Liu R, Holik AZ, Su S, Jansz N, Chen K, Leong HS, et al. Why weight? Modelling sample and observational level variability improves power in RNA-seq analyses. Nucleic Acids Res. 2015;43(15):e97. doi: 10.1093/nar/gkv412 25925576.

99. Phipson B, Lee S, Majewski IJ, Alexander WS, Smyth GK. Robust Hyperparameter Estimation Protects against Hypervariable Genes and Improves Power to Detect Differential Expression. Ann Appl Stat. 2016;10(2):946–63. doi: 10.1214/16-AOAS920 28367255.

100. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47. doi: 10.1093/nar/gkv007 25605792.

101. Risso D, Ngai J, Speed TP, Dudoit S. Normalization of RNA-seq data using factor analysis of control genes or samples. Nat Biotechnol. 2014;32(9):896–902. doi: 10.1038/nbt.2931 25150836.

102. Kolde R. pheatmap: Pretty Heatmaps. 1.0.8 ed2015.

103. Durinck S, Moreau Y, Kasprzyk A, Davis S, De Moor B, Brazma A, et al. BioMart and Bioconductor: a powerful link between biological databases and microarray data analysis. Bioinformatics. 2005;21(16):3439–40. doi: 10.1093/bioinformatics/bti525 16082012.

104. Durinck S, Spellman PT, Birney E, Huber W. Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt. Nat Protoc. 2009;4(8):1184–91. doi: 10.1038/nprot.2009.97 19617889.

105. Giner G, Smyth GK. FRY: a fast approximation to ROAST gene set test with mean aggregated set statistics [version 1; not peer reviewed]. F1000Research. 2016;5(2605). doi: 10.7490/f1000research.1113351.1

106. Wu D, Lim E, Vaillant F, Asselin-Labat ML, Visvader JE, Smyth GK. ROAST: rotation gene set tests for complex microarray experiments. Bioinformatics. 2010;26(17):2176–82. doi: 10.1093/bioinformatics/btq401 20610611.

107. Heinz S, Benner C, Spann N, Bertolino E, Lin YC, Laslo P, et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell. 2010;38(4):576–89. doi: 10.1016/j.molcel.2010.05.004 20513432.

108. Benner C. HOMER (Hypergeometric Optimization of Motif EnRichment). v4.9 ed2017. p. Software for motif discovery and next generation sequencing analysis.

109. Choi M, Chang CY, Clough T, Broudy D, Killeen T, MacLean B, et al. MSstats: an R package for statistical analysis of quantitative mass spectrometry-based proteomic experiments. Bioinformatics. 2014;30(17):2524–6. doi: 10.1093/bioinformatics/btu305 24794931.

110. Kammers K, Cole RN, Tiengwe C, Ruczinski I. Detecting Significant Changes in Protein Abundance. EuPA Open Proteom. 2015;7:11–9. doi: 10.1016/j.euprot.2015.02.002 25821719.

111. Magis AT, Funk CC, Price ND. SNAPR: a bioinformatics pipeline for efficient and accurate RNA-seq alignment and analysis. IEEE Life Sci Lett. 2015;1(2):22–5. Epub 2015/08/28. doi: 10.1109/LLS.2015.2465870 29270443.

112. Langfelder P, Zhang B, Horvath S. Defining clusters from a hierarchical cluster tree: the Dynamic Tree Cut package for R. Bioinformatics. 2008;24(5):719–20. doi: 10.1093/bioinformatics/btm563 18024473.

113. Langfelder P, Horvath S. Fast R Functions for Robust Correlations and Hierarchical Clustering. J Stat Softw. 2012;46(11). 23050260.

114. Allaire JJ, Gandrud C, Russell K, Yetman CJ. networkD3: D3 JavaScript Network Graphs from R. 2017.

115. Martin S, Brown WM, Klavans R, Boyack KW, editors. OpenOrd: an open-source toolbox for large graph layout. IS&T/SPIE Electronic Imaging; 2011: SPIE.

116. Kroehne V, Freudenreich D, Hans S, Kaslin J, Brand M. Regeneration of the adult zebrafish brain from neurogenic radial glia-type progenitors. Development. 2011:dev.072587. doi: 10.1242/dev.072587 22007133

117. Kaslin J, Kroehne V, Ganz J, Hans S, Brand M. Distinct roles of neuroepithelial-like and radial glia-like progenitor cells in cerebellar regeneration. Development. 2017;144(8):1462–71. Epub 2017/03/16. doi: 10.1242/dev.144907 28289134.


Článek vyšel v časopise

PLOS One


2020 Číslo 1
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

KOST
Koncepce osteologické péče pro gynekology a praktické lékaře
nový kurz
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Svět praktické medicíny 5/2023 (znalostní test z časopisu)

Imunopatologie? … a co my s tím???
Autoři: doc. MUDr. Helena Lahoda Brodská, Ph.D.

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#