Skip to main content

Advertisement

Log in

CRISPR, Prime Editing, Optogenetics, and DREADDs: New Therapeutic Approaches Provided by Emerging Technologies in the Treatment of Spinal Cord Injury

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Spinal cord injury (SCI) causes temporary disabilities or permanent effects including neuropathic pain and spastiscity. The damage often results from mechanical trauma, which in turn triggers the neuroinflammatory process. Neuroinflammation plays essential roles in the structural, biochemical, and cellular changes that take place in the spinal cord after the injury. Indeed, SCI activates many different signaling pathways that coordinate the resulting cellular responses. While neuroinflammation serves as a physiological reaction to harmful stimuli, it is clear that long-lasting inflammatory response leads to aggravation of the neurodegenerative processes, becoming detrimental to recovery post-injury. In this context, we present some possible therapeutic targets in these activated signaling pathways and provide new perspectives for SCI treatment based on recently developed technologies, including clustered regularly interspaced short palindromic repeats (CRISPR)-based methods (including prime editing), optogenetics, and designer receptor exclusively activated by designer drugs (DREADDs). We critically analyze the recent advances in the deployment of these methods focusing on the control of the initial neuroinflammatory response. We then propose alternatives and provide new avenues for SCI treatment based on these emerging technologies.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Singh A, Tetreault L, Kalsi-Ryan S, Nouri A, Fehlings MG (2014) Global prevalence and incidence of traumatic spinal cord injury. Clin Epidemiol 6:309–331. https://doi.org/10.2147/CLEP.S68889

    Article  PubMed  PubMed Central  Google Scholar 

  2. Kasinathan N, Vanathi MB, Subrahmanyam VM, Rao JV (2015) A review on response of immune system in spinal cord injury and therapeutic agents useful in treatment. Curr Pharm Biotechnol 16(1):26–34

    Article  CAS  Google Scholar 

  3. Thuret S, Moon LD, Gage FH (2006) Therapeutic interventions after spinal cord injury. Nat Rev Neurosci 7(8):628–643. https://doi.org/10.1038/nrn1955

    Article  CAS  PubMed  Google Scholar 

  4. Finnerup NB (2017) Neuropathic pain and spasticity: intricate consequences of spinal cord injury. Spinal Cord 55(12):1046–1050. https://doi.org/10.1038/sc.2017.70

    Article  CAS  PubMed  Google Scholar 

  5. Linn FH, Franssen H, Notermans NC, Ramos LM, Wokke JH (1994) Segmental fasciculations as a late sequel of spinal cord injury. Clin Neurol Neurosurg 96(2):174–177

    Article  CAS  Google Scholar 

  6. Alizadeh A, Dyck SM, Karimi-Abdolrezaee S (2019) Traumatic spinal cord injury: an overview of pathophysiology, models and acute injury mechanisms. Front Neurol 10:282. https://doi.org/10.3389/fneur.2019.00282

    Article  PubMed  PubMed Central  Google Scholar 

  7. Lude P, Kennedy P, Elfstrom ML, Ballert CS (2014) Quality of life in and after spinal cord injury rehabilitation: a longitudinal multicenter study. Top Spinal Cord Inj Rehabil 20(3):197–207. https://doi.org/10.1310/sci2003-197

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Huang H, Sun T, Chen L, Moviglia G, Chernykh E, von Wild K, Deda H, Kang KS et al (2014) Consensus of clinical neurorestorative progress in patients with complete chronic spinal cord injury. Cell Transplant 23(Suppl 1):S5–S17. https://doi.org/10.3727/096368914X684952

  9. Oyinbo CA (2011) Secondary injury mechanisms in traumatic spinal cord injury: a nugget of this multiply cascade. Acta Neurobiol Exp (Wars) 71(2):281–299

    Google Scholar 

  10. Paschon V, Higa GS, Walter LT, de Sousa E, Zuzarte FC, Weber VR, Resende RR, Kihara AH (2013) A new and reliable guide for studies of neuronal loss based on focal lesions and combinations of in vivo and in vitro approaches. PLoS One 8(4):e60486. https://doi.org/10.1371/journal.pone.0060486

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Fleming JC, Norenberg MD, Ramsay DA, Dekaban GA, Marcillo AE, Saenz AD, Pasquale-Styles M, Dietrich WD et al (2006) The cellular inflammatory response in human spinal cords after injury. Brain 129(Pt 12):3249–3269. https://doi.org/10.1093/brain/awl296

    Article  PubMed  Google Scholar 

  12. Forn-Cuni G, Varela M, Pereiro P, Novoa B, Figueras A (2017) Conserved gene regulation during acute inflammation between zebrafish and mammals. Sci Rep 7:41905. https://doi.org/10.1038/srep41905

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Bimbova K, Bacova M, Kisucka A, Pavel J, Galik J, Zavacky P, Marsala M, Stropkovska A et al (2018) A single dose of atorvastatin applied acutely after spinal cord injury suppresses inflammation, apoptosis, and promotes axon outgrowth, which might be essential for favorable functional outcome. Int J Mol Sci 19(4). https://doi.org/10.3390/ijms19041106

  14. Alastrue-Agudo A, Rodriguez-Jimenez FJ, Mocholi EL, De Giorgio F, Erceg S, Moreno-Manzano V (2018) FM19G11 and ependymal progenitor/stem cell combinatory treatment enhances neuronal preservation and oligodendrogenesis after severe spinal cord injury. Int J Mol Sci 19(1). https://doi.org/10.3390/ijms19010200

  15. Park J, Decker JT, Margul DJ, Smith DR, Cummings BJ, Anderson AJ, Shea LD (2018) Local immunomodulation with anti-inflammatory cytokine-encoding lentivirus enhances functional recovery after spinal cord injury. Mol Ther 26(7):1756–1770. https://doi.org/10.1016/j.ymthe.2018.04.022

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Yu CG, Yezierski RP (2005) Activation of the ERK1/2 signaling cascade by excitotoxic spinal cord injury. Brain Res Mol Brain Res 138(2):244–255. https://doi.org/10.1016/j.molbrainres.2005.04.013

    Article  CAS  PubMed  Google Scholar 

  17. Yu CG, Yezierski RP, Joshi A, Raza K, Li Y, Geddes JW (2010) Involvement of ERK2 in traumatic spinal cord injury. J Neurochem 113(1):131–142. https://doi.org/10.1111/j.1471-4159.2010.06579.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Fan B, Wei Z, Yao X, Shi G, Cheng X, Zhou X, Zhou H, Ning G et al (2018) Microenvironment imbalance of spinal cord injury. Cell Transplant 27(6):853–866. https://doi.org/10.1177/0963689718755778

    Article  PubMed  PubMed Central  Google Scholar 

  19. Covarrubias AJ, Aksoylar HI, Horng T (2015) Control of macrophage metabolism and activation by mTOR and Akt signaling. Semin Immunol 27(4):286–296. https://doi.org/10.1016/j.smim.2015.08.001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Troutman TD, Bazan JF, Pasare C (2012) Toll-like receptors, signaling adapters and regulation of the pro-inflammatory response by PI3K. Cell Cycle 11(19):3559–3567. https://doi.org/10.4161/cc.21572

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Polumuri SK, Toshchakov VY, Vogel SN (2007) Role of phosphatidylinositol-3 kinase in transcriptional regulation of TLR-induced IL-12 and IL-10 by Fc gamma receptor ligation in murine macrophages. J Immunol 179(1):236–246. https://doi.org/10.4049/jimmunol.179.1.236

    Article  CAS  PubMed  Google Scholar 

  22. Sutherland TC, Mathews KJ, Mao Y, Nguyen T, Gorrie CA (2016) Differences in the cellular response to acute spinal cord injury between developing and mature rats highlights the potential significance of the inflammatory response. Front Cell Neurosci 10:310. https://doi.org/10.3389/fncel.2016.00310

    Article  CAS  PubMed  Google Scholar 

  23. Raingeaud J, Whitmarsh AJ, Barrett T, Derijard B, Davis RJ (1996) MKK3- and MKK6-regulated gene expression is mediated by the p38 mitogen-activated protein kinase signal transduction pathway. Mol Cell Biol 16(3):1247–1255. https://doi.org/10.1128/mcb.16.3.1247

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chen J, Wang Q, Zhou W, Zhou Z, Tang PY, Xu T, Liu W, Li LW et al (2018) GPCR kinase 2-interacting protein-1 protects against ischemia-reperfusion injury of the spinal cord by modulating ASK1/JNK/p38 signaling. FASEB J:fj201800548. https://doi.org/10.1096/fj.201800548

  25. Eishingdrelo H, Kongsamut S (2013) Minireview: targeting GPCR activated ERK pathways for drug discovery. Curr Chem Genom Transl Med 7:9–15. https://doi.org/10.2174/2213988501307010009

    Article  PubMed  PubMed Central  Google Scholar 

  26. Cruz CD, Cruz F (2007) The ERK 1 and 2 pathway in the nervous system: from basic aspects to possible clinical applications in pain and visceral dysfunction. Curr Neuropharmacol 5(4):244–252. https://doi.org/10.2174/157015907782793630

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gopfert C, Andreas N (2018) The p38-MK2/3 module is critical for IL-33-induced signaling and cytokine production in dendritic cells. 200 (3):1198-1206. doi:https://doi.org/10.4049/jimmunol.1700727

  28. Guess AJ, Ayoob R, Chanley M, Manley J, Cajaiba MM, Agrawal S, Pengal R, Pyle AL et al (2013) Crucial roles of the protein kinases MK2 and MK3 in a mouse model of glomerulonephritis. PLoS One 8(1):e54239. https://doi.org/10.1371/journal.pone.0054239

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Roux PP, Blenis J (2004) ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev 68(2):320–344. https://doi.org/10.1128/mmbr.68.2.320-344.2004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Tchevkina E, Komelkov A (2012) Protein phosphorylation as a key mechanism of mTORC1/2 signaling pathways. In. https://doi.org/10.5772/48274

  31. Zaheer A, Lim R (1998) Overexpression of glia maturation factor (GMF) in PC12 pheochromocytoma cells activates p38 MAP kinase, MAPKAP kinase-2, and tyrosine hydroxylase. Biochem Biophys Res Commun 250(2):278–282. https://doi.org/10.1006/bbrc.1998.9301

    Article  CAS  PubMed  Google Scholar 

  32. Zhou ZW, Li XX, He ZX, Pan ST, Yang Y, Zhang X, Chow K, Yang T et al (2015) Induction of apoptosis and autophagy via sirtuin1- and PI3K/Akt/mTOR-mediated pathways by plumbagin in human prostate cancer cells. Drug Des Devel Ther 9:1511–1554. https://doi.org/10.2147/dddt.s75976

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kasuya Y, Umezawa H, Hatano M (2018) Stress-activated protein kinases in spinal cord injury: focus on roles of p38. Int J Mol Sci 19(3). https://doi.org/10.3390/ijms19030867

  34. Yang W, Guo Q, Cheng Z, Wang Y, Bai N, He Z (2019) mTOR signaling pathway of spinal cord is involved in peripheral nerve injury-induced hyperalgesia in rats. Zhong Nan Da Xue Xue Bao Yi Xue Ban 44(4):377–385. https://doi.org/10.11817/j.issn.1672-7347.2019.04.006

    Article  PubMed  Google Scholar 

  35. Zubilewicz A, Hecquet C, Jeanny J-C, Soubrane G, Courtois Y, Mascarelli F (2001) Proliferation of CECs requires dual signaling through both MAPK/ERK and PI 3-K/Akt pathways. Invest Ophthalmol Vis Sci 42(2):488–496

    CAS  PubMed  Google Scholar 

  36. Lu Z, Xu S (2006) ERK1/2 MAP kinases in cell survival and apoptosis. IUBMB Life 58(11):621–631. https://doi.org/10.1080/15216540600957438

    Article  CAS  PubMed  Google Scholar 

  37. Popiolek-Barczyk K, Mika J (2016) Targeting the microglial signaling pathways: new insights in the modulation of neuropathic pain. Curr Med Chem 23(26):2908–2928. https://doi.org/10.2174/0929867323666160607120124

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Vergadi E, Ieronymaki E, Lyroni K, Vaporidi K, Tsatsanis C (2017) Akt signaling pathway in macrophage activation and M1/M2 polarization. J Immunol 198(3):1006–1014. https://doi.org/10.4049/jimmunol.1601515

    Article  CAS  PubMed  Google Scholar 

  39. Rahimian R, Lively S, Abdelhamid E, Lalancette-Hebert M, Schlichter L, Sato S, Kriz J (2019) Delayed galectin-3-mediated reprogramming of microglia after stroke is protective. Mol Neurobiol 56(9):6371–6385. https://doi.org/10.1007/s12035-019-1527-0

    Article  CAS  PubMed  Google Scholar 

  40. Bhat SA, Sood A, Shukla R, Hanif K (2019) AT2R activation prevents microglia pro-inflammatory activation in a NOX-dependent manner: inhibition of PKC activation and p47(phox) phosphorylation by PP2A. Mol Neurobiol 56(4):3005–3023. https://doi.org/10.1007/s12035-018-1272-9

    Article  CAS  PubMed  Google Scholar 

  41. Zhang J, Zheng Y, Luo Y, Du Y, Zhang X, Fu J (2019) Curcumin inhibits LPS-induced neuroinflammation by promoting microglial M2 polarization via TREM2/ TLR4/ NF-kappaB pathways in BV2 cells. Mol Immunol 116:29–37. https://doi.org/10.1016/j.molimm.2019.09.020

    Article  CAS  PubMed  Google Scholar 

  42. Younger D, Murugan M, Rama Rao KV, Wu LJ, Chandra N (2019) Microglia receptors in animal models of traumatic brain injury. Mol Neurobiol 56(7):5202–5228. https://doi.org/10.1007/s12035-018-1428-7

    Article  CAS  PubMed  Google Scholar 

  43. Dong H, Zhang W, Zeng X, Hu G, Zhang H, He S, Zhang S (2014) Histamine induces upregulated expression of histamine receptors and increases release of inflammatory mediators from microglia. Mol Neurobiol 49(3):1487–1500. https://doi.org/10.1007/s12035-014-8697-6

    Article  CAS  PubMed  Google Scholar 

  44. Subramaniam SR, Federoff HJ (2017) Targeting microglial activation states as a therapeutic avenue in Parkinson’s disease. Front Aging Neurosci 9:176. https://doi.org/10.3389/fnagi.2017.00176

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ransohoff RM (2016) A polarizing question: do M1 and M2 microglia exist? Nat Neurosci 19(8):987–991. https://doi.org/10.1038/nn.4338

    Article  CAS  PubMed  Google Scholar 

  46. Koss K, Churchward MA, Tsui C, Todd KG (2019) In vitro priming and hyper-activation of brain microglia: an assessment of phenotypes. Mol Neurobiol 56(9):6409–6425. https://doi.org/10.1007/s12035-019-1529-y

    Article  CAS  PubMed  Google Scholar 

  47. Noristani HN, Gerber YN, Sabourin JC, Le Corre M, Lonjon N, Mestre-Frances N, Hirbec HE, Perrin FE (2017) RNA-Seq analysis of microglia reveals time-dependent activation of specific genetic programs following spinal cord injury. Front Mol Neurosci 10:90. https://doi.org/10.3389/fnmol.2017.00090

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Xu Z, Wang BR, Wang X, Kuang F, Duan XL, Jiao XY, Ju G (2006) ERK1/2 and p38 mitogen-activated protein kinase mediate iNOS-induced spinal neuron degeneration after acute traumatic spinal cord injury. Life Sci 79(20):1895–1905. https://doi.org/10.1016/j.lfs.2006.06.023

    Article  CAS  PubMed  Google Scholar 

  49. Dello Russo C, Lisi L, Tringali G, Navarra P (2009) Involvement of mTOR kinase in cytokine-dependent microglial activation and cell proliferation. Biochem Pharmacol 78(9):1242–1251. https://doi.org/10.1016/j.bcp.2009.06.097

    Article  CAS  Google Scholar 

  50. Lu DY, Liou HC, Tang CH, Fu WM (2006) Hypoxia-induced iNOS expression in microglia is regulated by the PI3-kinase/Akt/mTOR signaling pathway and activation of hypoxia inducible factor-1alpha. Biochem Pharmacol 72(8):992–1000. https://doi.org/10.1016/j.bcp.2006.06.038

    Article  CAS  PubMed  Google Scholar 

  51. Kanno H, Ozawa H, Sekiguchi A, Yamaya S, Tateda S, Yahata K, Itoi E (2012) The role of mTOR signaling pathway in spinal cord injury. Cell Cycle 11(17):3175–3179. https://doi.org/10.4161/cc.21262

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Perrin FE, Noristani HN (2019) Serotonergic mechanisms in spinal cord injury. Exp Neurol 318:174–191. https://doi.org/10.1016/j.expneurol.2019.05.007

    Article  CAS  PubMed  Google Scholar 

  53. Fan B, Sun Y-J, Liu S-Y, Che L, Li G-Y (2017) Neuroprotective strategy in retinal degeneration: suppressing ER stress-induced cell death via inhibition of the mTOR signal. Int J Mol Sci 18(1):201. https://doi.org/10.3390/ijms18010201

    Article  CAS  PubMed Central  Google Scholar 

  54. Grasso S, Piedad M, Carrasco-Garca E, Mayor-Lpez L, Tristante E, Rocamora L, Gmez-Martnez n, Garca-Morales P, A J, Saceda M, Martnez-Lacaci I (2012) Cell death and cancer, Novel Therapeutic Strategies In doi:https://doi.org/10.5772/51285

  55. Ouyang W, Li J, Ma Q, Huang C (2006) Essential roles of PI-3K/Akt/IKKbeta/NFkappaB pathway in cyclin D1 induction by arsenite in JB6 Cl41 cells. Carcinogenesis 27(4):864–873. https://doi.org/10.1093/carcin/bgi321

    Article  CAS  PubMed  Google Scholar 

  56. Zhang L, Huang C, Guo Y, Gou X (2015) MicroRNA-26b modulates the NF-kappaB pathway in alveolar macrophages by regulating PTEN. 195 (11):5404-5414. doi:https://doi.org/10.4049/jimmunol.1402933

  57. McGonnell IM, Grigoriadis AE, Lam EW, Price JS, Sunters A (2012) A specific role for phosphoinositide 3-kinase and AKT in osteoblasts? Front Endocrinol (Lausanne) 3:88. https://doi.org/10.3389/fendo.2012.00088

    Article  Google Scholar 

  58. Grilo AL, Mantalaris A (2019) Apoptosis: a mammalian cell bioprocessing perspective. Biotechnol Adv 37(3):459–475. https://doi.org/10.1016/j.biotechadv.2019.02.012

    Article  CAS  PubMed  Google Scholar 

  59. Georgescu MM (2010) PTEN tumor suppressor network in PI3K-Akt pathway control. Genes Cancer 1(12):1170–1177. https://doi.org/10.1177/1947601911407325

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Song Z, Han X, Shen L, Zou H, Zhang B, Liu J, Gong A (2018) PTEN silencing enhances neuronal proliferation and differentiation by activating PI3K/Akt/GSK3beta pathway in vitro. Exp Cell Res 363(2):179–187. https://doi.org/10.1016/j.yexcr.2018.01.001

    Article  CAS  PubMed  Google Scholar 

  61. Liu T, Cao FJ, Xu DD, Xu YQ, Feng SQ (2015) Upregulated Ras/Raf/ERK1/2 signaling pathway: a new hope in the repair of spinal cord injury. Neural Regen Res 10(5):792–796. https://doi.org/10.4103/1673-5374.156984

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Zhang P, Zhang L, Zhu L, Chen F, Zhou S, Tian T, Zhang Y, Jiang X et al (2015) The change tendency of PI3K/Akt pathway after spinal cord injury. Am J Transl Res 7(11):2223–2232

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Hahn-Windgassen A, Nogueira V, Chen CC, Skeen JE, Sonenberg N, Hay N (2005) Akt activates the mammalian target of rapamycin by regulating cellular ATP level and AMPK activity. J Biol Chem 280(37):32081–32089. https://doi.org/10.1074/jbc.M502876200

    Article  CAS  PubMed  Google Scholar 

  64. Park KK, Liu K, Hu Y, Smith PD, Wang C, Cai B, Xu B, Connolly L et al (2008) Promoting axon regeneration in the adult CNS by modulation of the PTEN/mTOR pathway. Science 322(5903):963–966. https://doi.org/10.1126/science.1161566

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Liu K, Lu Y, Lee JK, Samara R, Willenberg R, Sears-Kraxberger I, Tedeschi A, Park KK et al (2010) PTEN deletion enhances the regenerative ability of adult corticospinal neurons. Nat Neurosci 13(9):1075–1081. https://doi.org/10.1038/nn.2603

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Park KK, Liu K, Hu Y, Kanter JL, He Z (2010) PTEN/mTOR and axon regeneration. Exp Neurol 223(1):45–50. https://doi.org/10.1016/j.expneurol.2009.12.032

    Article  CAS  PubMed  Google Scholar 

  67. Ning K, Drepper C, Valori CF, Ahsan M, Wyles M, Higginbottom A, Herrmann T, Shaw P et al (2010) PTEN depletion rescues axonal growth defect and improves survival in SMN-deficient motor neurons. Hum Mol Genet 19(16):3159–3168. https://doi.org/10.1093/hmg/ddq226

    Article  CAS  PubMed  Google Scholar 

  68. Ohtake Y, Park D, Abdul-Muneer PM, Li H, Xu B, Sharma K, Smith GM, Selzer ME et al (2014) The effect of systemic PTEN antagonist peptides on axon growth and functional recovery after spinal cord injury. Biomaterials 35(16):4610–4626. https://doi.org/10.1016/j.biomaterials.2014.02.037

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Gerber YN, Saint-Martin GP, Bringuier CM, Bartolami S, Goze-Bac C, Noristani HN, Perrin FE (2018) CSF1R inhibition reduces microglia proliferation, promotes tissue preservation and improves motor recovery after spinal cord injury. Front Cell Neurosci 12:368. https://doi.org/10.3389/fncel.2018.00368

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Teixeira WGJ, Cristante AF, Marcon RM, Bispo G, Ferreira R, de Barros-Filho TEP (2018) Granulocyte colony-stimulating factor combined with methylprednisolone improves functional outcomes in rats with experimental acute spinal cord injury. Clinics (Sao Paulo) 73:e235. https://doi.org/10.6061/clinics/2018/e235

    Article  Google Scholar 

  71. Matsuda S, Nakagawa Y, Tsuji A, Kitagishi Y, Nakanishi A, Murai T (2018) Implications of PI3K/AKT/PTEN signaling on superoxide dismutases expression and in the pathogenesis of Alzheimer’s disease. Diseases 6(2). https://doi.org/10.3390/diseases6020028

  72. Sanchez-Alegria K, Flores-Leon M, Avila-Munoz E, Rodriguez-Corona N, Arias C (2018) PI3K signaling in neurons: a central node for the control of multiple functions. Int J Mol Sci 19(12). https://doi.org/10.3390/ijms19123725

  73. Shi J, Yu J, Zhang Y, Wu L, Dong S, Wu L, Wu L, Du S et al (2019) PI3K/Akt pathway-mediated HO-1 induction regulates mitochondrial quality control and attenuates endotoxin-induced acute lung injury. Lab Investig. https://doi.org/10.1038/s41374-019-0286-x

  74. Rohlenova K, Neuzil J, Rohlena J (2016) The role of Her2 and other oncogenes of the PI3K/AKT pathway in mitochondria. Biol Chem 397(7):607–615. https://doi.org/10.1515/hsz-2016-0130

    Article  CAS  PubMed  Google Scholar 

  75. Paschon V, Morena BC, Correia FF, Beltrame GR, Dos Santos GB, Cristante AF, Kihara AH (2019) VDAC1 is essential for neurite maintenance and the inhibition of its oligomerization protects spinal cord from demyelination and facilitates locomotor function recovery after spinal cord injury. Sci Rep 9(1):14063. https://doi.org/10.1038/s41598-019-50506-4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Jiang Z, Zhang Y, Chen X, Lam PY, Yang H, Xu Q, Yu AC (2002) Activation of Erk1/2 and Akt in astrocytes under ischemia. Biochem Biophys Res Commun 294(3):726–733. https://doi.org/10.1016/S0006-291X(02)00540-5

    Article  CAS  PubMed  Google Scholar 

  77. Povysheva TV, Mukhamedshina YO, Rizvanov AA, Chelyshev YA (2018) PTEN expression in astrocytic processes after spinal cord injury. Mol Cell Neurosci 88:231–239. https://doi.org/10.1016/j.mcn.2018.02.008

    Article  CAS  PubMed  Google Scholar 

  78. Lian H, Yang L, Cole A, Sun L, Chiang AC, Fowler SW, Shim DJ, Rodriguez-Rivera J et al (2015) NFkappaB-activated astroglial release of complement C3 compromises neuronal morphology and function associated with Alzheimer’s disease. Neuron 85(1):101–115. https://doi.org/10.1016/j.neuron.2014.11.018

    Article  CAS  PubMed  Google Scholar 

  79. Ahn JY (2014) Neuroprotection signaling of nuclear akt in neuronal cells. Exp Neurobiol 23(3):200–206. https://doi.org/10.5607/en.2014.23.3.200

    Article  PubMed  PubMed Central  Google Scholar 

  80. Danilov CA, Steward O (2015) Conditional genetic deletion of PTEN after a spinal cord injury enhances regenerative growth of CST axons and motor function recovery in mice. Exp Neurol 266:147–160. https://doi.org/10.1016/j.expneurol.2015.02.012

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Liu SQ, Zhang ML, Zhang HJ, Liu FZ, Chu RJ, Zhang GX, Zhu L (2017) Matrine promotes oligodendrocyte development in CNS autoimmunity through the PI3K/Akt signaling pathway. Life Sci 180:36–41. https://doi.org/10.1016/j.lfs.2017.05.010

    Article  CAS  PubMed  Google Scholar 

  82. Flores AI, Mallon BS, Matsui T, Ogawa W, Rosenzweig A, Okamoto T, Macklin WB (2000) Akt-mediated survival of oligodendrocytes induced by neuregulins. J Neurosci 20(20):7622–7630

    Article  CAS  Google Scholar 

  83. Gaesser JM, Fyffe-Maricich SL (2016) Intracellular signaling pathway regulation of myelination and remyelination in the CNS. Exp Neurol 283(Pt B):501–511. https://doi.org/10.1016/j.expneurol.2016.03.008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Lu H, Zhang LH, Yang L, Tang PF (2018) The PI3K/Akt/FOXO3a pathway regulates regeneration following spinal cord injury in adult rats through TNF-alpha and p27kip1 expression. Int J Mol Med 41(5):2832–2838. https://doi.org/10.3892/ijmm.2018.3459

    Article  CAS  PubMed  Google Scholar 

  85. Medina I, Friedel P, Rivera C, Kahle KT, Kourdougli N, Uvarov P, Pellegrino C (2014) Current view on the functional regulation of the neuronal K(+)-Cl(-) cotransporter KCC2. Front Cell Neurosci 8:27. https://doi.org/10.3389/fncel.2014.00027

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Chen B, Li Y, Yu B, Zhang Z, Brommer B, Williams PR, Liu Y, Hegarty SV et al (2018) Reactivation of dormant relay pathways in injured spinal cord by KCC2 manipulations. Cell 174(6):1599. https://doi.org/10.1016/j.cell.2018.08.050

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Gompf HS, Budygin EA, Fuller PM, Bass CE (2015) Targeted genetic manipulations of neuronal subtypes using promoter-specific combinatorial AAVs in wild-type animals. Front Behav Neurosci 9:152. https://doi.org/10.3389/fnbeh.2015.00152

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Zheng C, Baum BJ (2008) Evaluation of promoters for use in tissue-specific gene delivery. Methods Mol Biol 434:205–219. https://doi.org/10.1007/978-1-60327-248-3_13

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Brown AJ, James DC (2017) Constructing strong cell type-specific promoters through informed design. Methods Mol Biol 1651:131–145. https://doi.org/10.1007/978-1-4939-7223-4_10

    Article  CAS  PubMed  Google Scholar 

  90. deHaseth PL, Zupancic ML, Record MT Jr (1998) RNA polymerase-promoter interactions: the comings and goings of RNA polymerase. J Bacteriol 180(12):3019–3025

    Article  CAS  Google Scholar 

  91. Walther W, Stein U (1996) Cell type specific and inducible promoters for vectors in gene therapy as an approach for cell targeting. J Mol Med (Berl) 74(7):379–392

    Article  CAS  Google Scholar 

  92. Wu SH, Liao ZX, DR J, Zheng N, Zhang LH, Tang H, He XB, Wu Y et al (2017) Comparative study of the transfection efficiency of commonly used viral vectors in rhesus monkey (Macaca mulatta) brains. Zool Res 38(2):88–95. https://doi.org/10.24272/j.issn.2095-8137.2017.015

    Article  PubMed  PubMed Central  Google Scholar 

  93. Buchholz CJ, Friedel T, Buning H (2015) Surface-engineered viral vectors for selective and cell type-specific gene delivery. Trends Biotechnol 33(12):777–790. https://doi.org/10.1016/j.tibtech.2015.09.008

    Article  CAS  PubMed  Google Scholar 

  94. Nguyen DN, Green JJ, Chan JM, Longer R, Anderson DG (2009) Polymeric materials for gene delivery and DNA vaccination. Adv Mater 21(8):847–867. https://doi.org/10.1002/adma.200801478

    Article  CAS  PubMed  Google Scholar 

  95. Green JJ, Shi J, Chiu E, Leshchiner ES, Langer R, Anderson DG (2006) Biodegradable polymeric vectors for gene delivery to human endothelial cells. Bioconjug Chem 17(5):1162–1169. https://doi.org/10.1021/bc0600968

    Article  CAS  PubMed  Google Scholar 

  96. Pannier AK, Shea LD (2004) Controlled release systems for DNA delivery. Mol Ther 10(1):19–26. https://doi.org/10.1016/j.ymthe.2004.03.020

    Article  CAS  PubMed  Google Scholar 

  97. Agbay A, Edgar JM, Robinson M, Styan T, Wilson K, Schroll J, Ko J, Khadem Mohtaram N et al (2016) Biomaterial strategies for delivering stem cells as a treatment for spinal cord injury. Cells Tissues Organs 202(1-2):42–51. https://doi.org/10.1159/000446474

    Article  CAS  PubMed  Google Scholar 

  98. Edgar JM, Robinson M, Willerth SM (2017) Fibrin hydrogels induce mixed dorsal/ventral spinal neuron identities during differentiation of human induced pluripotent stem cells. Acta Biomater 51:237–245. https://doi.org/10.1016/j.actbio.2017.01.040

    Article  CAS  PubMed  Google Scholar 

  99. Robinson M, Douglas S, Michelle Willerth S (2017) Mechanically stable fibrin scaffolds promote viability and induce neurite outgrowth in neural aggregates derived from human induced pluripotent stem cells. Sci Rep 7(1):6250. https://doi.org/10.1038/s41598-017-06570-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Wang F, Wang L, Zou X, Duan S, Li Z, Deng Z, Luo J, Lee SY et al (2019) Advances in CRISPR-Cas systems for RNA targeting, tracking and editing. Biotechnol Adv 37(5):708–729. https://doi.org/10.1016/j.biotechadv.2019.03.016

    Article  CAS  PubMed  Google Scholar 

  101. Mali P, Yang L, Esvelt KM, Aach J, Guell M, DiCarlo JE, Norville JE, Church GM (2013) RNA-guided human genome engineering via Cas9. Science 339(6121):823–826. https://doi.org/10.1126/science.1232033

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Tian X, Gu T, Patel S, Bode AM, Lee MH, Dong Z (2019) CRISPR/Cas9 -an evolving biological tool kit for cancer biology and oncology. NPJ Precis Oncol 3:8. https://doi.org/10.1038/s41698-019-0080-7

    Article  PubMed  PubMed Central  Google Scholar 

  103. Zischewski J, Fischer R, Bortesi L (2017) Detection of on-target and off-target mutations generated by CRISPR/Cas9 and other sequence-specific nucleases. Biotechnol Adv 35(1):95–104. https://doi.org/10.1016/j.biotechadv.2016.12.003

    Article  CAS  PubMed  Google Scholar 

  104. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E (2012) A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337(6096):816–821. https://doi.org/10.1126/science.1225829

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Bortesi L, Fischer R (2015) The CRISPR/Cas9 system for plant genome editing and beyond. Biotechnol Adv 33(1):41–52. https://doi.org/10.1016/j.biotechadv.2014.12.006

    Article  CAS  PubMed  Google Scholar 

  106. Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD (2010) Genome editing with engineered zinc finger nucleases. Nat Rev Genet 11(9):636–646. https://doi.org/10.1038/nrg2842

    Article  CAS  PubMed  Google Scholar 

  107. Makarova KS, Zhang F, Koonin EV (2017) SnapShot: class 2 CRISPR-Cas systems. Cell 168(1):328–328.e321. https://doi.org/10.1016/j.cell.2016.12.038

  108. Pickar-Oliver A, Gersbach CA (2019) The next generation of CRISPR–Cas technologies and applications. Nat Rev Mol Cell Biol 20(8):490–507. https://doi.org/10.1038/s41580-019-0131-5

    Article  CAS  PubMed  Google Scholar 

  109. Swarts DC, van der Oost J, Jinek M (2017) Structural basis for guide RNA processing and seed-dependent DNA targeting by CRISPR-Cas12a. Mol Cell 66(2):221–233 e224. https://doi.org/10.1016/j.molcel.2017.03.016

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Yan M-Y, Yan H-Q, Ren G-X, Zhao J-P, Guo X-P, Sun Y-C (2017) CRISPR-Cas12a-assisted recombineering in bacteria. Appl Environ Microbiol 83(17):e00947–e00917. https://doi.org/10.1128/AEM.00947-17

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Yao R, Liu D, Jia X, Zheng Y, Liu W, Xiao Y (2018) CRISPR-Cas9/Cas12a biotechnology and application in bacteria. Synth Syst Biotechnol 3(3):135–149. https://doi.org/10.1016/j.synbio.2018.09.004

    Article  PubMed  PubMed Central  Google Scholar 

  112. Cohen J (2019) Prime editing promises to be a cut above CRISPR. Science 366(6464):406. https://doi.org/10.1126/science.366.6464.406

    Article  CAS  PubMed  Google Scholar 

  113. Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C et al (2019) Search-and-replace genome editing without double-strand breaks or donor DNA. Nature. https://doi.org/10.1038/s41586-019-1711-4

  114. Blasi E, Barluzzi R, Bocchini V, Mazzolla R, Bistoni F (1990) Immortalization of murine microglial cells by a v-raf/v-myc carrying retrovirus. J Neuroimmunol 27(2-3):229–237. https://doi.org/10.1016/0165-5728(90)90073-v

    Article  CAS  Google Scholar 

  115. Raikwar SP, Thangavel R, Dubova I, Selvakumar GP, Ahmed ME, Kempuraj D, Zaheer SA, Iyer SS et al (2019) Targeted gene editing of glia maturation factor in microglia: a novel Alzheimer’s disease therapeutic target. Mol Neurobiol 56(1):378–393. https://doi.org/10.1007/s12035-018-1068-y

    Article  CAS  PubMed  Google Scholar 

  116. Guo Q, Li S, Liang Y, Zhang Y, Zhang J, Wen C, Lin S, Wang H et al (2010) Effects of C3 deficiency on inflammation and regeneration following spinal cord injury in mice. Neurosci Lett 485(1):32–36. https://doi.org/10.1016/j.neulet.2010.08.056

    Article  CAS  PubMed  Google Scholar 

  117. Zhang W, Wang G, Wang Y, Jin Y, Zhao L, Xiong Q, Zhang L, Mou L et al (2017) Generation of complement protein C3 deficient pigs by CRISPR/Cas9-mediated gene targeting. Sci Rep 7(1):5009. https://doi.org/10.1038/s41598-017-05400-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Savell KE, Day JJ (2017) Applications of CRISPR/Cas9 in the mammalian central nervous system. Yale J Biol Med 90(4):567–581

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Straub C, Granger AJ, Saulnier JL, Sabatini BL (2014) CRISPR/Cas9-mediated gene knock-down in post-mitotic neurons. PLoS One 9(8):e105584. https://doi.org/10.1371/journal.pone.0105584

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Staahl BT, Benekareddy M, Coulon-Bainier C, Banfal AA, Floor SN, Sabo JK, Urnes C, Munares GA et al (2017) Efficient genome editing in the mouse brain by local delivery of engineered Cas9 ribonucleoprotein complexes. Nat Biotechnol 35(5):431–434. https://doi.org/10.1038/nbt.3806

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Zheng Y, Shen W, Zhang J, Yang B, Liu YN, Qi H, Yu X, Lu SY et al (2018) Author correction: CRISPR interference-based specific and efficient gene inactivation in the brain. Nat Neurosci 21(6):894. https://doi.org/10.1038/s41593-018-0125-1

    Article  CAS  PubMed  Google Scholar 

  122. Liu J, Gao C, Chen W, Ma W, Li X, Shi Y, Zhang H, Zhang L et al (2016) CRISPR/Cas9 facilitates investigation of neural circuit disease using human iPSCs: mechanism of epilepsy caused by an SCN1A loss-of-function mutation. Transl Psychiatry 6:e703. https://doi.org/10.1038/tp.2015.203

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Savell KE, Bach SV, Zipperly ME, Revanna JS, Goska NA, Tuscher JJ, Duke CG, Sultan FA et al (2019) A neuron-optimized CRISPR/dCas9 activation system for robust and specific gene regulation. eNeuro 6(1). https://doi.org/10.1523/ENEURO.0495-18.2019

  124. Callif BL, Maunze B, Krueger NL, Simpson MT, Blackmore MG (2017) The application of CRISPR technology to high content screening in primary neurons. Mol Cell Neurosci 80:170–179. https://doi.org/10.1016/j.mcn.2017.01.003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Maeder ML, Stefanidakis M, Wilson CJ, Baral R, Barrera LA, Bounoutas GS, Bumcrot D, Chao H et al (2019) Development of a gene-editing approach to restore vision loss in Leber congenital amaurosis type 10. Nat Med 25(2):229–233. https://doi.org/10.1038/s41591-018-0327-9

    Article  CAS  PubMed  Google Scholar 

  126. Hodges CA, Conlon RA (2019) Delivering on the promise of gene editing for cystic fibrosis. Genes Diseases 6(2):97–108. https://doi.org/10.1016/j.gendis.2018.11.005

    Article  CAS  PubMed  Google Scholar 

  127. Fenno L, Yizhar O, Deisseroth K (2011) The development and application of optogenetics. Annu Rev Neurosci 34:389–412. https://doi.org/10.1146/annurev-neuro-061010-113817

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Mahmoudi P, Veladi H, Pakdel FG (2017) Optogenetics, tools and applications in neurobiology. J Med Signals Sens 7(2):71–79

    Article  Google Scholar 

  129. Bass CE, Grinevich VP, Gioia D, Day-Brown JD, Bonin KD, Stuber GD, Weiner JL, Budygin EA (2013) Optogenetic stimulation of VTA dopamine neurons reveals that tonic but not phasic patterns of dopamine transmission reduce ethanol self-administration. Front Behav Neurosci 7:173. https://doi.org/10.3389/fnbeh.2013.00173

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Wiegert JS, Mahn M, Prigge M, Printz Y, Yizhar O (2017) Silencing neurons: tools, applications, and experimental constraints. Neuron 95(3):504–529. https://doi.org/10.1016/j.neuron.2017.06.050

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Wietek J, Wiegert JS, Adeishvili N, Schneider F, Watanabe H, Tsunoda SP, Vogt A, Elstner M et al (2014) Conversion of channelrhodopsin into a light-gated chloride channel. Science 344(6182):409–412. https://doi.org/10.1126/science.1249375

    Article  CAS  PubMed  Google Scholar 

  132. Fortin DL, Dunn TW, Fedorchak A, Allen D, Montpetit R, Banghart MR, Trauner D, Adelman JP et al (2011) Optogenetic photochemical control of designer K+ channels in mammalian neurons. J Neurophysiol 106(1):488–496. https://doi.org/10.1152/jn.00251.2011

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Toettcher JE, Weiner OD, Lim WA (2013) Using optogenetics to interrogate the dynamic control of signal transmission by the Ras/Erk module. Cell 155(6):1422–1434. https://doi.org/10.1016/j.cell.2013.11.004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Hughes RM, Freeman DJ, Lamb KN, Pollet RM, Smith WJ, Lawrence DS (2015) Optogenetic apoptosis: light-triggered cell death. Angew Chem Int Ed Eng 54(41):12064–12068. https://doi.org/10.1002/anie.201506346

    Article  CAS  Google Scholar 

  135. Jewhurst K, Levin M, McLaughlin KA (2014) Optogenetic control of apoptosis in targeted tissues of Xenopus laevis embryos. J Cell Death 7:25–31. https://doi.org/10.4137/JCD.S18368

    Article  PubMed  PubMed Central  Google Scholar 

  136. Ono K, Suzuki H, Yamamoto R, Sahashi H, Takido Y, Sawada M (2017) Optogenetic control of cell differentiation in channelrhodopsin-2-expressing OS3, a bipotential glial progenitor cell line. Neurochem Int 104:49–63. https://doi.org/10.1016/j.neuint.2016.12.022

    Article  CAS  PubMed  Google Scholar 

  137. Sokolik C, Liu Y, Bauer D, McPherson J, Broeker M, Heimberg G, Qi LS, Sivak DA et al (2015) Transcription factor competition allows embryonic stem cells to distinguish authentic signals from noise. Cell Syst 1(2):117–129. https://doi.org/10.1016/j.cels.2015.08.001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Zhang K, Cui B (2015) Optogenetic control of intracellular signaling pathways. Trends Biotechnol 33(2):92–100. https://doi.org/10.1016/j.tibtech.2014.11.007

    Article  CAS  PubMed  Google Scholar 

  139. Ghazale H, Ripoll C, Leventoux N, Jacob L, Azar S, Mamaeva D, Glasson Y, Calvo CF et al (2019) RNA profiling of the human and mouse spinal cord stem cell niches reveals an embryonic-like regionalization with MSX1(+) roof-plate-derived cells. Stem Cell Reports 12(5):1159–1177. https://doi.org/10.1016/j.stemcr.2019.04.001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Daadi MM, Klausner JQ, Bajar B, Goshen I, Lee-Messer C, Lee SY, Winge MC, Ramakrishnan C et al (2016) Optogenetic stimulation of neural grafts enhances neurotransmission and downregulates the inflammatory response in experimental stroke model. Cell Transplant 25(7):1371–1380. https://doi.org/10.3727/096368915X688533

    Article  PubMed  Google Scholar 

  141. Houle JD, Tom VJ, Mayes D, Wagoner G, Phillips N, Silver J (2006) Combining an autologous peripheral nervous system “bridge” and matrix modification by chondroitinase allows robust, functional regeneration beyond a hemisection lesion of the adult rat spinal cord. J Neurosci 26(28):7405–7415. https://doi.org/10.1523/JNEUROSCI.1166-06.2006

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Ahmad A, Ashraf S, Komai S (2015) Optogenetics applications for treating spinal cord injury. Asian Spine J 9(2):299–305. https://doi.org/10.4184/asj.2015.9.2.299

    Article  PubMed  PubMed Central  Google Scholar 

  143. Airan RD, Thompson KR, Fenno LE, Bernstein H, Deisseroth K (2009) Temporally precise in vivo control of intracellular signalling. Nature 458(7241):1025–1029. https://doi.org/10.1038/nature07926

    Article  CAS  PubMed  Google Scholar 

  144. English JD, Sweatt JD (1996) Activation of p42 mitogen-activated protein kinase in hippocampal long term potentiation. J Biol Chem 271(40):24329–24332. https://doi.org/10.1074/jbc.271.40.24329

    Article  CAS  PubMed  Google Scholar 

  145. English JD, Sweatt JD (1997) A requirement for the mitogen-activated protein kinase cascade in hippocampal long term potentiation. J Biol Chem 272(31):19103–19106. https://doi.org/10.1074/jbc.272.31.19103

    Article  CAS  PubMed  Google Scholar 

  146. Winder DG, Martin KC, Muzzio IA, Rohrer D, Chruscinski A, Kobilka B, Kandel ER (1999) ERK plays a regulatory role in induction of LTP by theta frequency stimulation and its modulation by beta-adrenergic receptors. Neuron 24(3):715–726

    Article  CAS  Google Scholar 

  147. Kyung T, Lee S, Kim JE, Cho T, Park H, Jeong YM, Kim D, Shin A et al (2015) Optogenetic control of endogenous Ca(2+) channels in vivo. Nat Biotechnol 33(10):1092–1096. https://doi.org/10.1038/nbt.3350

    Article  CAS  PubMed  Google Scholar 

  148. Munaron L (2002) Calcium signalling and control of cell proliferation by tyrosine kinase receptors (review). Int J Mol Med 10(6):671–676

    CAS  PubMed  Google Scholar 

  149. Chierzi S, Ratto GM, Verma P, Fawcett JW (2005) The ability of axons to regenerate their growth cones depends on axonal type and age, and is regulated by calcium, cAMP and ERK. Eur J Neurosci 21(8):2051–2062. https://doi.org/10.1111/j.1460-9568.2005.04066.x

    Article  PubMed  Google Scholar 

  150. Herberts CA, Kwa MS, Hermsen HP (2011) Risk factors in the development of stem cell therapy. J Transl Med 9:29. https://doi.org/10.1186/1479-5876-9-29

    Article  PubMed  PubMed Central  Google Scholar 

  151. Vieira MS, Santos AK, Vasconcellos R, Goulart VAM, Parreira RC, Kihara AH, Ulrich H, Resende RR (2018) Neural stem cell differentiation into mature neurons: mechanisms of regulation and biotechnological applications. Biotechnol Adv 36(7):1946–1970. https://doi.org/10.1016/j.biotechadv.2018.08.002

    Article  CAS  PubMed  Google Scholar 

  152. Shao J, Wang M, Yu G, Zhu S, Yu Y, Heng BC, Wu J, Ye H (2018) Synthetic far-red light-mediated CRISPR-dCas9 device for inducing functional neuronal differentiation. Proc Natl Acad Sci U S A 115(29):E6722–E6730. https://doi.org/10.1073/pnas.1802448115

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Lee HM, Giguere PM, Roth BL (2014) DREADDs: novel tools for drug discovery and development. Drug Discov Today 19(4):469–473. https://doi.org/10.1016/j.drudis.2013.10.018

    Article  CAS  PubMed  Google Scholar 

  154. Rogan SC, Roth BL (2011) Remote control of neuronal signaling. Pharmacol Rev 63(2):291–315. https://doi.org/10.1124/pr.110.003020

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Armbruster BN, Li X, Pausch MH, Herlitze S, Roth BL (2007) Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand. Proc Natl Acad Sci U S A 104(12):5163–5168. https://doi.org/10.1073/pnas.0700293104

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Smith KS, Bucci DJ, Luikart BW, Mahler SV (2016) DREADDS: use and application in behavioral neuroscience. Behav Neurosci 130(2):137–155. https://doi.org/10.1037/bne0000135

    Article  PubMed  PubMed Central  Google Scholar 

  157. Zhu H, Roth BL (2014) DREADD: a chemogenetic GPCR signaling platform. Int J Neuropsychopharmacol 18(1). https://doi.org/10.1093/ijnp/pyu007

  158. Grace PM, Wang X, Strand KA, Baratta MV, Zhang Y, Galer EL, Yin H, Maier SF et al (2018) DREADDed microglia in pain: Implications for spinal inflammatory signaling in male rats. Exp Neurol 304:125–131. https://doi.org/10.1016/j.expneurol.2018.03.005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Wang Z, Maunze B, Wang Y, Tsoulfas P, Blackmore MG (2018) Global connectivity and function of descending spinal input revealed by 3D microscopy and retrograde transduction. J Neurosci 38(49):10566–10581. https://doi.org/10.1523/JNEUROSCI.1196-18.2018

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Jaiswal PB, Mistretta OC, Ward PJ, English AW (2018) Chemogenetic enhancement of axon regeneration following peripheral nerve injury in the SLICK-A mouse. Brain Sci 8(5). https://doi.org/10.3390/brainsci8050093

  161. Blazquez C, Chiarlone A, Bellocchio L, Resel E, Pruunsild P, Garcia-Rincon D, Sendtner M, Timmusk T et al (2015) The CB(1) cannabinoid receptor signals striatal neuroprotection via a PI3K/Akt/mTORC1/BDNF pathway. Cell Death Differ 22(10):1618–1629. https://doi.org/10.1038/cdd.2015.11

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Mitew S, Gobius I, Fenlon LR, McDougall SJ, Hawkes D, Xing YL, Bujalka H, Gundlach AL et al (2018) Pharmacogenetic stimulation of neuronal activity increases myelination in an axon-specific manner. Nat Commun 9(1):306. https://doi.org/10.1038/s41467-017-02719-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Snaidero N, Mobius W, Czopka T, Hekking LH, Mathisen C, Verkleij D, Goebbels S, Edgar J et al (2014) Myelin membrane wrapping of CNS axons by PI(3,4,5)P3-dependent polarized growth at the inner tongue. Cell 156(1-2):277–290. https://doi.org/10.1016/j.cell.2013.11.044

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Bedbrook CN, Deverman BE, Gradinaru V (2018) Viral strategies for targeting the central and peripheral nervous systems. Annu Rev Neurosci 41:323–348. https://doi.org/10.1146/annurev-neuro-080317-062048

    Article  CAS  PubMed  Google Scholar 

  165. Cyranoski D (2016) CRISPR gene-editing tested in a person for the first time. Nature 539(7630):479. https://doi.org/10.1038/nature.2016.20988

    Article  CAS  PubMed  Google Scholar 

  166. Cyranoski D (2016) Chinese scientists to pioneer first human CRISPR trial. Nature 535(7613):476–477. https://doi.org/10.1038/nature.2016.20302

    Article  CAS  PubMed  Google Scholar 

  167. You L, Tong R, Li M, Liu Y, Xue J, Lu Y (2019) Advancements and obstacles of CRISPR-Cas9 technology in translational research. Mol Ther Methods Clin Dev 13:359–370. https://doi.org/10.1016/j.omtm.2019.02.008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Bi A, Cui J, Ma YP, Olshevskaya E, Pu M, Dizhoor AM, Pan ZH (2006) Ectopic expression of a microbial-type rhodopsin restores visual responses in mice with photoreceptor degeneration. Neuron 50(1):23–33. https://doi.org/10.1016/j.neuron.2006.02.026

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Szablowski JO, Lee-Gosselin A, Lue B, Malounda D, Shapiro MG (2018) Acoustically targeted chemogenetics for the non-invasive control of neural circuits. Nat Biomed Eng 2(7):475–484. https://doi.org/10.1038/s41551-018-0258-2

    Article  CAS  PubMed  Google Scholar 

  170. Chen Q, Zeng Z, Hu Z (2012) Optogenetics in neuroscience: what we gain from studies in mammals. Neurosci Bull 28(4):423–434. https://doi.org/10.1007/s12264-012-1250-6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Roth BL (2016) DREADDs for neuroscientists. Neuron 89(4):683–694. https://doi.org/10.1016/j.neuron.2016.01.040

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Ordaz J, Wu W, Xu X-M (2017) Optogenetics and its application in neural degeneration and regeneration. Neural Regen Res 12(8):1197–1209. https://doi.org/10.4103/1673-5374.213532

    Article  PubMed  PubMed Central  Google Scholar 

  173. Vann KT, Xiong Z-G (2016) Optogenetics for neurodegenerative diseases. Int J Physiol Pathophysiol Pharmacol 8(1):1–8

    PubMed  PubMed Central  Google Scholar 

  174. Bryson JB, Machado CB, Lieberam I, Greensmith L (2016) Restoring motor function using optogenetics and neural engraftment. Curr Opin Biotechnol 40:75–81. https://doi.org/10.1016/j.copbio.2016.02.016

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank Silva Honda Takada, Erika Reime Kinjo, Guilherme Shigueto Villar Higa, Mariana Sacrini A. Ferraz, and Fernando da Silva Borges for scientific discussions.

Funding

This work was financially supported by grants from Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP, no. 2017/26439-0), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq, no. 431000/2016-6), and research scholarships from Universidade Federal do ABC (UFABC). Dr. Willerth receives funding from NSERC, the Canada Research Chairs program, and the Alzheimer’s Association.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Alexandre Fogaça Cristante, Stephanie Michelle Willerth, Florence Evelyne Perrin or Alexandre Hiroaki Kihara.

Ethics declarations

Disclaimer

The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Paschon, V., Correia, F.F., Morena, B.C. et al. CRISPR, Prime Editing, Optogenetics, and DREADDs: New Therapeutic Approaches Provided by Emerging Technologies in the Treatment of Spinal Cord Injury. Mol Neurobiol 57, 2085–2100 (2020). https://doi.org/10.1007/s12035-019-01861-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-019-01861-w

Keywords

Navigation