Introduction
The golden era of biotechnology ushered in a plethora of new tools for our toolkits, including precision gene editing via homologous recombination [
1], base editing[
2], prime editing[
3,
4,
5], large serine recombinase-mediate gene insertion[
6], and CRISPR transposases[
7]. These have the potential to cure any genetic illness. With adeno-associated viral (AAV) vectors, gene therapy has been brought to bear in the human body, sometimes greatly improving function in patients[
8]. However, full cures are generally not possible, and many genetic diseases remain untreatable currently. In the cases wherein effective treatments or cures are not attainable, it is not for a lack of in vitro tools, but rather our ability to deliver them to the cells around the body that need to be repaired.
With regard to central nervous system (CNS) disorders, AAV.CAP-Mac was recently described, which can cross the blood-brain barrier and reach cells throughout the brain[
9]. However, although parts of the cortex and thalamus were effectively transduced in 17-year-old adult rhesus monkeys, other regions of the brain were not[
9]. It is also possible that AAV.CAP-Mac may not work in humans as well as it did in non-human primates. Additionally, the cost of producing sufficiently high titers of AAV vectors and other viral vectors for therapeutic purposes is at least currently very steep[
10,
11,
12]. Moreover, AAV therapies are possibly genotoxic or cytotoxic, especially at high doses[
13]. In contrast to AAV vectors, a transient, off-the-shelf, cell-based treatment enabled by inducible mRNA delivery would likely be much cheaper and safer.
Microglial Replacement
The small molecule CSF1R inhibitor, PLX5622, potently depletes microglia[
14]. PLX3397 is another, FDA-approved small molecule CSF1R inhibitor, and it also potently depletes microglia in mice and non-human primates[
15]. Microglial depletion in both model organisms appears safe, and can be repeated multiple times in mice, given sufficient time between depletion-repopulation cycles[
16,
17].
PLX3397 may also potently deplete microglia in humans as well, although the clinical trial that measured microglial depletion may not have administered PLX3397 for long enough to thoroughly test its efficacy in this regard[
18]. It might be possible to edit induced pluripotent stem cell-derived macrophages (iPSC-Macs)
ex vivo, and then infuse them intrathecally or intravenously to replace a patient's microglia with ones that can employ a system called COURIER (controlled output and uptake of RNA for interrogation, expression, and regulation)[
19,
20].
Normally, the remaining population of microglia or peripheral monocytes might outcompete iPSC-Macs that are infused into the bloodstream[
21,
22], but an inhibitor-resistant CSF1R variant has been developed[
23]. Thus, constant selection is possible - which should enable non-invasive microglial replacement.
For gene editing purposes, the iPSC-Macs could at least eventually be “off-the-shelf”[
24,
25]. This would substantially decrease the cost of the therapy. After treatment, a small molecule like a rapamycin analog that can penetrate the blood-brain barrier (BBB) can be administered to eliminate them via caspase-9 activation[
26], and the patient’s hematopoietic stem cells (HSCs) would repopulate the microglia.
Non-genotoxic HSC transplant is also a possibility if the direct intravenous or intrathecal infusion of edited cells is ineffective[
27].
Also, while microglia sample their surrounding microenvironment constantly with protrusions that extend and retract, they may not move around much from place to place[
28,
29]. To address this potential issue, random migration of the iPSC-Mac-derived microglia in the CNS could be induced or enhanced by inhibiting LRRK2[
30]. Other methods of inducing hyper-motility are also possible[
31,
32].
Neuronal Gene Editing
Base editing of the SMN2 gene in neurons is a viable strategy for spinal muscular atrophy[
33,
34]. In this case, the COURIER cargo could be an mRNA molecule encoding a zinc finger base editor. However, they currently have more off-target activity than CRISPR base editors[
35]. TALE base editors exist, as well[
36]. Alternatively, the cargo could be a self-amplifying RNA (saRNA) vector[
37]. An saRNA vector could enable the use of a CRISPR base editor, wherein a subgenomic promoter effectively replicates an sgRNA module but weakly replicates the proteinaceous component of the base editor[
38,
39]. As larger RNA molecules were packaged less efficiently in COURIER, but dual delivery was possible, a trans-amplifying RNA (taRNA) vector could be employed[
40]. In either case, the RNA-dependent RNA polymerase could be inhibited in the edited microglia via an orthogonal degree, perhaps.
With saRNA and taRNA vectors, there may be some considerations with superinfection exclusion, copy number restriction, and possibly gene dosage compensation[
41,
42,
43].
As opposed to an AAV vector, microglial replacement and cell-based treatment might be too slow for SMA1 patients. However, as described in[
33], clinicians could co-administer the antisense oligonucleotide drug nusinersen to extend the therapeutic window.
Two other CNS genetic illnesses that microglial replacement could potentially help with are Tay-Sachs and Huntington’s disease. For Tay-Sachs, prime editing could remove a causative four-base duplication[
3]. Base editing could diminish the disease-causing mutation in Huntington’s[
44]. Twin prime editing combined with a site-specific recombinase could also enable the targeted excision of large trinucleotide repeat regions[
4,
45].
Furthermore, the APOE4 allele, which is a genetic risk factor for Alzheimer’s disease, could be edited to APOE3[
46].
Secreted Particle Information Transfer (SPIT) could also be exported for gene editing[
47]
. However, mRNA export may be more efficacious, as opposed to RNP secretion. Also, SPIT utilizes viral capsid proteins, which would probably be potently immunogenic.
COURIER Immunogenicity
As Dr. Horns et al. mentioned[
20], COURIER protein nanocages may not stimulate an excessive immune response. Low-dose dexamethasone could possibly be sufficient to prevent undue inflammation from all of the COURIER components. Dexamethasone has been used to counter excessive immune responses in patients with SARS-CoV-2[
48].
If not, there are other strategies. The innate immune response to dsRNA generated from vector replication could possibly be attenuated by the expression of the MERS-CoV ORFa protein[
49]. If cyclic induction is required over a long period of time, the adaptive immune response to the gene editing components may need to be attenuated as well. First, the vector could encode a deimmunized dCas9 protein[
50]. Second, it may help if the vector were to incorporate multiple, tandem miR-142-3p binding sites in the dCas9 mRNA 3’UTR[
51]. Third, the vector could express the SARS-CoV-2 ORF6 protein, which inhibits the MHC class I pathway[
52].
Of course, adding more elements requires a more packaging space. If necessary, one could theoretically minimize the size of the required elements. Or, as was also mentioned in the COURIER article, alternative nanocage architectures could tune cargo capacity.
Other Uses for Microglial Replacement
CNS Senescent Cell Elimination
Senescent cells in the CNS could be eliminated by a system involving a replicating RNA COURIER cargo molecule and ADAR-mediated detection of the p16Ink4a transcript[
53,
54]
. Detection would lead to the triggering of caspase 8 production.
The caspase’s activity could be attenuated via mutation if necessary to ensure a proper therapeutic window.
Mitochondrial Transplantation
Instead of COURIER, arrestin domain containing protein 1 [ARRDC1]-mediated microvesicles (ARMMs) could be employed to transfer mitochondria to aged cells with mutated or damaged mitochondria[
55,
56]
.
PGC-1α could be overexpressed by the edited microglia to increase their intracellular stores of mitochondria[
57]
.
TFAM could also be directly overexpressed in order to maximally increase the mitochondrial DNA (mtDNA) copy number of the pristine mtDNA in the edited microglia. However, PGC-1α overexpression would accomplish this to some extent at least.
Miro1 overexpression might help to improve donation efficiency[
58]
.
These mitochondria could be imbued with SNPs in non-coding regions - or synonymous or non-synonymous SNPs - that allow for targeting of aged mtDNA with pre-imported nucleases[
59]
. Alternatively, the ARMMs could be loaded with mRNA encoding nucleases. This might work better than pre-imported nucleases because it would not rely on target cell mitochondrial fusion-fission dynamics. COURIER can be used instead of loading the mRNA into ARMMs, but would not be directional as currently formulated.
Some SNPs are actually associated with endurance-enhancing or pro-longevity effects [
60,
61]
.
Perhaps an ideal approach would be to load saRNA or taRNA into the ARMMs that encode TFAM and the nucleases - to destroy old mtDNA, while simultaneously promoting the replication of the new mtDNA.
Conclusion
Microglial replacement in combination with COURIER or SPIT could enable cheap and effective CNS gene editing.
References
- Nambiar TS, Billon P, Diedenhofen G, Hayward SB, Taglialatela A, Cai K, et al. Stimulation of CRISPR-mediated homology-directed repair by an engineered RAD18 variant. Nat Commun 2019;10:3395. [CrossRef]
- Davis JR, Wang X, Witte IP, Huang TP, Levy JM, Raguram A, et al. Efficient in vivo base editing via single adeno-associated viruses with size-optimized genomes encoding compact adenine base editors. Nat Biomed Eng 2022;6:1272–83. [CrossRef]
- Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019;576:149–57. [CrossRef]
- Anzalone AV, Gao XD, Podracky CJ, Nelson AT, Koblan LW, Raguram A, et al. Programmable deletion, replacement, integration and inversion of large DNA sequences with twin prime editing. Nat Biotechnol 2022;40:731–40. [CrossRef]
- Davis JR, Banskota S, Levy JM, Newby GA, Wang X, Anzalone AV, et al. Efficient prime editing in mouse brain, liver and heart with dual AAVs. Nat Biotechnol 2023:1–12. [CrossRef]
- Durrant MG, Fanton A, Tycko J, Hinks M, Chandrasekaran SS, Perry NT, et al. Systematic discovery of recombinases for efficient integration of large DNA sequences into the human genome. Nat Biotechnol 2022:1–12. [CrossRef]
- Tou CJ, Orr B, Kleinstiver BP. Precise cut-and-paste DNA insertion using engineered type V-K CRISPR-associated transposases. Nat Biotechnol 2023;41:968–79. [CrossRef]
- Day JW, Finkel RS, Chiriboga CA, Connolly AM, Crawford TO, Darras BT, et al. Onasemnogene abeparvovec gene therapy for symptomatic infantile-onset spinal muscular atrophy in patients with two copies of SMN2 (STR1VE): an open-label, single-arm, multicentre, phase 3 trial. The Lancet Neurology 2021;20:284–93. [CrossRef]
- Chuapoco MR, Flytzanis NC, Goeden N, et al. Adeno-associated viral vectors for functional intravenous gene transfer throughout the non-human primate brain. Nat Nanotechnol 2023;1–11. [CrossRef]
- Collins LT, Ponnazhagan S, Curiel DT. Synthetic Biology Design as a Paradigm Shift toward Manufacturing Affordable Adeno-Associated Virus Gene Therapies. ACS Synth Biol 2023;12:17–26. [CrossRef]
- Becker Z. Sporting a $3.5M price tag, CSL and uniQure’s hemophilia B gene therapy crosses FDA finish line. Fierce Pharma 2022. https://www.fiercepharma.com/pharma/csl-and-uniqures-hemophilia-b-gene-therapy-scores-approval-35-million-price-tag (accessed December 9, 2023).
- Bluebird Bio Secures Deal with Large Commercial Payer for Lyfgenia Amid Price Concerns. BioSpace n.d. https://www.biospace.com/article/bluebird-bio-secures-deal-with-large-commercial-payer-for-lyfgenia-amid-price-concerns/ (accessed January 4, 2024).
- Davé UP, Cornetta K. AAV Joins the Rank of Genotoxic Vectors. Molecular Therapy 2021;29(2):418–419. [CrossRef]
- Spangenberg E, Severson PL, Hohsfield LA, et al. Sustained microglial depletion with CSF1R inhibitor impairs parenchymal plaque development in an Alzheimer’s disease model. Nat Commun 2019;10(1):3758. [CrossRef]
- Green KN, Crapser JD, Hohsfield LA. To Kill a Microglia: A Case for CSF1R Inhibitors. Trends in Immunology 2020;41(9):771–784. [CrossRef]
- Hillmer AT, Holden D, Fowles K, et al. Microglial depletion and activation: A [11C]PBR28 PET study in nonhuman primates. EJNMMI Res 2017;7:59. [CrossRef]
- Najafi AR, Crapser J, Jiang S, et al. A limited capacity for microglial repopulation in the adult brain. Glia 2018;66(11):2385–2396. [CrossRef]
- Butowski N, Colman H, De Groot JF, et al. Orally administered colony stimulating factor 1 receptor inhibitor PLX3397 in recurrent glioblastoma: an Ivy Foundation Early Phase Clinical Trials Consortium phase II study. Neuro Oncol 2016;18(4):557–564. [CrossRef]
- Ackermann M, Rafiei Hashtchin A, Manstein F, Carvalho Oliveira M, Kempf H, Zweigerdt R, et al. Continuous human iPSC-macrophage production by suspension culture in stirred tank bioreactors. Nat Protoc 2022;17:513–39. [CrossRef]
- Horns F, Martinez JA, Fan C, et al. Engineering RNA export for measurement and manipulation of living cells. Cell 2023;186(17):3642-3658.e32; [CrossRef]
- Cronk JC, Filiano AJ, Louveau A, Marin I, Marsh R, Ji E, et al. Peripherally derived macrophages can engraft the brain independent of irradiation and maintain an identity distinct from microglia. J Exp Med 2018;215:1627–47. [CrossRef]
- Lund H, Pieber M, Parsa R, Han J, Grommisch D, Ewing E, et al. Competitive repopulation of an empty microglial niche yields functionally distinct subsets of microglia-like cells. Nat Commun 2018;9:4845. [CrossRef]
- Chadarevian JP, Lombroso SI, Peet GC, Hasselmann J, Tu C, Marzan DE, et al. Engineering an inhibitor-resistant human CSF1R variant for microglia replacement. Journal of Experimental Medicine 2022;220:e20220857. [CrossRef]
- Jo S, Das S, Williams A, et al. Endowing universal CAR T-cell with immune-evasive properties using TALEN-gene editing. Nat Commun 2022;13:3453. [CrossRef]
- Rao Y, Peng B. Allogenic microglia replacement: A novel therapeutic strategy for neurological disorders. Fundamental Research 2024;4(2):237–245. [CrossRef]
- Stavrou M, Philip B, Traynor-White C, Davis CG, Onuoha S, Cordoba S, et al. A Rapamycin-Activated Caspase 9-Based Suicide Gene. Mol Ther 2018;26:1266–76. [CrossRef]
- Hayal TB, Wu C, Abraham D, et al. The Impact of CD45-Antibody-Drug Conjugate Conditioning on Clonal Dynamics and Immune Tolerance Post HSPC Transplantation in Rhesus Macaques. Blood 2023;142:3419. [CrossRef]
- Nimmerjahn A, Kirchhoff F, Helmchen F. Resting Microglial Cells Are Highly Dynamic Surveillants of Brain Parenchyma in Vivo. Science 2005;308:1314–8. [CrossRef]
- Zhang Y, Wei D, Wang X, Wang B, Li M, Fang H, et al. Run-and-Tumble Dynamics and Mechanotaxis Discovered in Microglial Migration. Research 2023;6:0063. [CrossRef]
- Choi I, Kim B, Byun J-W, Baik SH, Huh YH, Kim J-H, et al. LRRK2 G2019S mutation attenuates microglial motility by inhibiting focal adhesion kinase. Nat Commun 2015;6:8255. [CrossRef]
- Tabdanov ED, Rodríguez-Merced NJ, Cartagena-Rivera AX, et al. Engineering T cells to enhance 3D migration through structurally and mechanically complex tumor microenvironments. Nat Commun 2021;12(1):2815. [CrossRef]
- Insall RH, Paschke P, Tweedy L. Steering yourself by the bootstraps: how cells create their own gradients for chemotaxis. Trends in Cell Biology 2022;32(7):585–596. [CrossRef]
- Arbab M, Matuszek Z, Kray KM, Du A, Newby GA, Blatnik AJ, et al. Base editing rescue of spinal muscular atrophy in cells and in mice. Science 2023;380:eadg6518. [CrossRef]
- Alves CRR, Ha LL, Yaworski R, Sutton ER, Lazzarotto CR, Christie KA, et al. Optimization of base editors for the functional correction of SMN2 as a treatment for spinal muscular atrophy. Nat Biomed Eng 2023:1–14. [CrossRef]
- Willis JCW, Silva-Pinheiro P, Widdup L, Minczuk M, Liu DR. Compact zinc finger base editors that edit mitochondrial or nuclear DNA in vitro and in vivo. Nat Commun 2022;13:7204. [CrossRef]
- Mok BY, Kotrys AV, Raguram A, Huang TP, Mootha VK, Liu DR. CRISPR-free base editors with enhanced activity and expanded targeting scope in mitochondrial and nuclear DNA. Nat Biotechnol 2022;40:1378–87. [CrossRef]
- Mc Cafferty S, De Temmerman J, Kitada T, Becraft JR, Weiss R, Irvine DJ, et al. In Vivo Validation of a Reversible Small Molecule-Based Switch for Synthetic Self-Amplifying mRNA Regulation. Molecular Therapy 2021;29:1164–73. [CrossRef]
- Lee RTH, Ng ASM, Ingham PW. Ribozyme Mediated gRNA Generation for In Vitro and In Vivo CRISPR/Cas9 Mutagenesis. PLOS ONE 2016;11:e0166020. [CrossRef]
- Oh Y, Kim H, Lee H-J, Kim S-G. Ribozyme-processed guide RNA enhances virus-mediated plant genome editing. Biotechnology Journal 2022;17:2100189. [CrossRef]
- Perkovic M, Gawletta S, Hempel T, Brill S, Nett E, Sahin U, et al. A trans-amplifying RNA simplified to essential elements is highly replicative and robustly immunogenic in mice. Mol Ther 2023;31:1636–46. [CrossRef]
- Reitmayer CM, Levitt E, Basu S, et al. Mimicking superinfection exclusion disrupts alphavirus infection and transmission in the yellow fever mosquito Aedes aegypti. Proceedings of the National Academy of Sciences 2023;120(37):e2303080120. [CrossRef]
- Li Z, Nagy PD. Diverse roles of host RNA binding proteins in RNA virus replication. RNA Biology 2011;8(2):305–315. [CrossRef]
- Yang J, Lee J, Land MA, et al. A synthetic circuit for buffering gene dosage variation between individual mammalian cells. Nat Commun 2021;12(1):4132. [CrossRef]
- Choi DE, Shin JW, Zeng S, Hong EP, Jang J-H, Loupe JM, et al. Base editing strategies to convert CAG to CAA diminish the disease-causing mutation in Huntington’s disease. eLife 2023;12. [CrossRef]
- Doman JL, Pandey S, Neugebauer ME, et al. Phage-assisted evolution and protein engineering yield compact, efficient prime editors. Cell 2023;186(18):3983-4002.e26. [CrossRef]
- Teter B, Campagna J, Zhu C, et al. Successful Gene Editing of Apolipoprotein E4 to E3 in Brain of Alzheimer Model Mice After a Single IV Dose of Synthetic Exosome-Delivered CRISPR. 2024;2024.04.23.590784. [CrossRef]
- Charlesworth CT, Homma S, Suchy F, et al. Secreted Particle Information Transfer (SPIT) – A Cellular Platform for In Vivo Genetic Engineering. bioRxiv 2024;2024.01.11.575257. [CrossRef]
- Wu H, Daouk S, Kebbe J, Chaudry F, Harper J, Brown B. Low-dose versus high-dose dexamethasone for hospitalized patients with COVID-19 pneumonia: A randomized clinical trial. PLoS One 2022;17:e0275217. [CrossRef]
- Blakney AK, McKay PF, Bouton CR, Hu K, Samnuan K, Shattock RJ. Innate Inhibiting Proteins Enhance Expression and Immunogenicity of Self-Amplifying RNA. Molecular Therapy 2021;29:1174–85. [CrossRef]
- Ferdosi SR, Ewaisha R, Moghadam F, Krishna S, Park JG, Ebrahimkhani MR, et al. Multifunctional CRISPR-Cas9 with engineered immunosilenced human T cell epitopes. Nat Commun 2019;10:1842. [CrossRef]
- Xiao Y, Muhuri M, Li S, Qin W, Xu G, Luo L, et al. Circumventing cellular immunity by miR142-mediated regulation sufficiently supports rAAV-delivered OVA expression without activating humoral immunity. JCI Insight n.d.;4:e99052. [CrossRef]
- Yoo J-S, Sasaki M, Cho SX, Kasuga Y, Zhu B, Ouda R, et al. SARS-CoV-2 inhibits induction of the MHC class I pathway by targeting the STAT1-IRF1-NLRC5 axis. Nat Commun 2021;12:6602. [CrossRef]
- Bussian TJ, Aziz A, Meyer CF, et al. Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 2018;562(7728):578–582. [CrossRef]
- Kaseniit KE, Katz N, Kolber NS, et al. Modular, programmable RNA sensing using ADAR editing in living cells. Nat Biotechnol 2023;41(4):482–487. [CrossRef]
- Phinney DG, Di Giuseppe M, Njah J, et al. Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nat Commun 2015;6:8472. [CrossRef]
- Wang Q, Yu J, Kadungure T, et al. ARMMs as a versatile platform for intracellular delivery of macromolecules. Nat Commun 2018;9(1):960. [CrossRef]
- Ventura-Clapier R, Garnier A, Veksler V. Transcriptional control of mitochondrial biogenesis: the central role of PGC-1α. Cardiovascular Research 2008;79(2):208–217. [CrossRef]
- Ahmad T, Mukherjee S, Pattnaik B, et al. Miro1 regulates intercellular mitochondrial transport & enhances mesenchymal stem cell rescue efficacy. The EMBO Journal 2014;33(9):994–1010. [CrossRef]
- Shoop WK, Lape J, Trum M, et al. Efficient elimination of MELAS-associated m.3243G mutant mitochondrial DNA by an engineered mitoARCUS nuclease. Nat Metab 2023;5(12):2169–2183. [CrossRef]
- Murakami H, Ota A, Simojo H, et al. Polymorphisms in Control Region of mtDNA Relates to Individual Differences in Endurance Capacity or Trainability. The Japanese Journal of Physiology 2002;52(3):247–256. [CrossRef]
- Castañeda V, Haro-Vinueza A, Salinas I, et al. The MitoAging Project: Single nucleotide polymorphisms (SNPs) in mitochondrial genes and their association to longevity. Mitochondrion 2022;66:13–26. [CrossRef]
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).