Abstract

Review Article

Vigour of CRISPR/Cas9 Gene Editing in Alzheimer’s Disease

Jes Paul*

Published: 05 October, 2018 | Volume 2 - Issue 2 | Pages: 047-051

Ailment repairing regiments has turn out to be arduous, despite a plenty of understanding and knowledge acquired in the past relating to the molecular underpinnings of Alzheimer’s disease (AD. Umpteen clinical experiments targeting the fabrication and accumulation have been turned fruitless to fit potency standards. The tests aiming beta-amyloid hypothesis also turned futile making it exigent for further handling tactics. The new emanation of a comparably candid, economical, and punctilious system known as gene editing have showed light in path of cure for AD by CRISPR/Cas9 gene editing. Being a straight approach this procedure has already shown assurance in other neurological disorders too such as Huntington’s disease. This review standpoint the immanent service of CRISPR/Cas9 as a remedial option for AD by aiming on specific genes inclusive of those that induce early-onset AD, as well as those that are substantial risk components for late-onset AD such as the apolipoprotein E4 (APOE4) gene.

Read Full Article HTML DOI: 10.29328/journal.jnnd.1001014 Cite this Article Read Full Article PDF

Keywords:

Alzheimer’s disease; CRISPR/Cas9; Gene editing; Treatment; Huntington’s disease; iPSC neurons

References

  1. Jones EL, Kalaria RN, Sharp SI, O’Brien JT, Francis PT, et al. Genetic associations of autopsy-confirmed vascular dementia subtypes. Dement GeriatrCognDisord. 2011; 31: 247–253. Ref.: https://goo.gl/VrBuco
  2. Mirra SS, Heyman A, McKeel D, Sumi SM, Crain BJ, et al. The consortium to establish a registry for Alzheimer’s disease (CERAD) part II. Standardization of the neuropathological assessment of Alzheimer’s disease. Neurol. 1991; 41: 479–486. Ref.: https://goo.gl/GZ4PKL
  3. Terry RD, Masliah E, Salmon DP, Butters N, DeTeresa R, et al. Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol. 1991; 30: 572–580. Ref.: https://goo.gl/UBTCiK
  4. DeKosky ST, Scheff SW, Styren SD. Structural correlates of cognition in dementia: Quantification and assessment of synapse change. Neurodegeneration. 1996; 5: 417–421. Ref.: https://goo.gl/vxUsux
  5. Coleman PD, Yao PJ. Synaptic slaughter in Alzheimer’s disease. Neurobiol Aging. 2003; 24: 1023–1027. Ref.: https://goo.gl/nezMzY
  6. Lacor PN, Buniel MC, Chang L, Fernandez SJ, Gong Y, et al. Synaptic targeting by Alzheimer’s-related amyloid beta oligomers. J Neurosci. 2004; 24: 10191–10200. Ref.: https://goo.gl/st1b1F
  7. Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, et al. Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci U S A. 1998; 95: 6448–6453. Ref.: https://goo.gl/wyxW1e
  8. McLean CA, Cherny RA, Fraser FW, Fuller SJ, Smith MJ, et al. Soluble pool of Abeta amyloid as a determinant of severity of neurodegeneration in Alzheimer’s disease. Ann Neurol. 1999; 46: 860–866. Ref.: https://goo.gl/ETDLih
  9. Broersen K, Rousseau F, Schymkowitz J. The culprit behind amyloid beta peptide related neurotoxicity in Alzheimer’s disease: Oligomer size or conformation? Alzheimers Res Ther. 2010; 2: 12. Ref.: https://goo.gl/sSVGiS
  10. Rohn TT, Kim N, Isho NF, Mack JM. The Potential of CRISPR/Cas9 Gene Editing as a Treatment Strategy for Alzheimer’s Disease. J Alzheimers Dis Parkinsonism. 2018; 8: 439. Ref.: https://goo.gl/7NDf8r
  11. Mojica FJ, Diez-Villasenor C, Garcia-Martinez J, Soria E. Intervening sequences of regularly spaced prokaryotic repeats derive from foreign genetic elements. J MolEvol. 2005; 60: 174–182. Ref.: https://goo.gl/oKLqZy
  12. Pourcel C, Salvignol G, Vergnaud G. CRISPR elements in Yersinia pestis acquire new repeats by preferential uptake of bacteriophage DNA, and provide additional tools for evolutionary studies. Microbiology. 2005; 151: 653–663. Ref.: https://goo.gl/wpw3tY
  13. Barrangou R, Fremaux C, Deveau H, Richards M, Boyaval P, et al. CRISPR provides acquired resistance against viruses in prokaryotes. Science. 2007; 315: 1709–1712. Ref.: https://goo.gl/jRYzbU
  14. Bolotin A, Quinquis B, Sorokin A, Ehrlich SD. Clustered regularly interspaced short palindrome repeats (CRISPRs) have spacers of extrachromosomal origin. Microbiology. 2005; 151: 2551–2561. Ref.: https://goo.gl/j9tdyj
  15. Deltcheva E, Chylinski K, Sharma CM, Gonzales K, Chao Y, et al. CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature. 2011; 471: 602–607. Ref.: https://goo.gl/LgTULX
  16. Saudou F, Humbert S. The biology of huntingtin. Neuron. 2016; 89: 910–926. Ref.: https://goo.gl/WGjZZv
  17. Bates GP, Dorsey R, Gusella JF, Hayden MR, Kay C, et al. Huntington disease. Nat Rev Dis Primers. 2015; 1: 15005. Ref.: https://goo.gl/sYTJU7
  18. Dabrowska M, Juzwa W, Krzyzosiak WJ, Olejniczak M. Precise excision of the CAG tract from the huntingtin gene by cas9 nickases. Front Neurosci. 2018; 12: 75. Ref.: https://goo.gl/juHxQe
  19. Schellenberg GD, Bird TD, Wijsman EM, Orr HT, Anderson L, et al. Genetic linkage evidence for a familial Alzheimer’s disease locus on chromosome 14. Science. 1992; 258: 668–671. Ref.: https://goo.gl/q2EmRY
  20. Levy-Lahad E, Wijsman EM, Nemens E, Anderson L, Goddard KA, et al. A familial Alzheimer’s disease locus on chromosome 1. Science. 1995; 269: 970–973. Ref.: https://goo.gl/2SYDSQ
  21. Vetrivel KS, Zhang YW, Xu H, Thinakaran G. Pathological and physiological functions of presenilins. MolNeurodegener. 2006; 1: 4. Ref.: https://goo.gl/Dbk6wU
  22. Pires C, Schmid B, Petræus C, Poon A, Nimsanor N, et al. Generation of a gene-corrected isogenic control cell line from an Alzheimer’s disease patient iPSC line carrying a A79V mutation in PSEN1. Stem Cell Res. 2016; 17: 285–288. Ref.: https://goo.gl/Bns14i
  23. Poon A, Schmid B, Pires C, Nielsen TT, Hjermind LE, et al. Generation of a gene-corrected isogenic control hiPSC line derived from a familial Alzheimer’s disease patient carrying a L150P mutation in presenilin 1. Stem Cell Res. 2016; 17: 466–469. Ref.: https://goo.gl/TAsC6J
  24. György B, Lööv C, Zaborowski M, Takeda S, Kleinstiver B, et al. CRISPR/Cas9 mediated disruption of the swedish APP allele as a therapeutic approach for early-onset alzheimer’s disease. MolTher Nucleic Acids. 2018; 11: 429–440. Ref.: https://goo.gl/5Q2R3R
  25. Sun J, Carlson-Stevermer J, Das U, Shen M, Delenclos M, et al. A CRISPR/Cas9 based strategy to manipulate the Alzheimer’s amyloid pathway. 2018 bioRxiv. Ref.: https://goo.gl/3NdTq9
  26. Das U, Scott DA, Ganguly A, Koo EH, Tang Y, et al. Activity-induced convergence of APP and BACE-1 in acidic microdomains via an endocytosis-dependent pathway. Neuron. 2013; 79: 447–460. Ref.: https://goo.gl/Q3nKnq
  27. Eisenstein M. Genetics: Finding risk factors. Nature. 2011; 475: S20–22. Ref.: https://goo.gl/GNjWRL
  28. Weisgraber KH, Rall SC, Jr, Mahley RW. Human E apoprotein heterogeneity. Cysteine-arginine interchanges in the amino acid sequence of the apo-E isoforms. J Biol Chem. 1981; 256: 9077–9083. Ref.: https://goo.gl/dn7yof
  29. Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and alzheimer disease meta-analysis consortium. JAMA. 1997; 278: 1349–1356. Ref.: https://goo.gl/VTAWzn
  30. Saunders AM, Strittmatter WJ, Schmechel D, George-Hyslop PH, Pericak-Vance MA, et al. Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer’s disease. Neurol. 1993; 43: 1467–1472. Ref.: https://goo.gl/nTC61T
  31. Kanekiyo T, Xu H, Bu G. ApoE and Aβ in Alzheimer’s disease: accidental encounters or partners? Neuron. 2014; 81: 740–754. Ref.: https://goo.gl/vuR6oA
  32. Wang C, Najm R, Xu Q, Jeong DE, Walker D, et al. Gain of toxic apolipoprotein E4 effects in human iPSC-derived neurons is ameliorated by a small-molecule structure corrector. Nat Med. 2018; 24: 647–657. Ref.: https://goo.gl/rvcjWZ
  33. Dong LM, Weisgraber KH. Human apolipoprotein E4 domain interaction. Arginine 61 and glutamic acid 255 interact to direct the preference for very low density lipoproteins. J Biol Chem. 1996; 271: 19053–19057. Ref.: https://goo.gl/bff8mX
  34. Prince M, Comas-Herrera A, Knapp M, Guerchet M, Karagiannidou M. Coverage, quality and costs now and in the future. Alzheimer’s Disease International; London: 2016. World Alzheimer Report 2016. Improving healthcare for people living with dementia.

Similar Articles

Recently Viewed

Read More

Most Viewed

Read More

Help ?