4.7 Article

CRISPR-Cas9 Mediated Exonic Disruption for HIV-1 Elimination

Journal

EBIOMEDICINE
Volume 73, Issue -, Pages -

Publisher

ELSEVIER
DOI: 10.1016/j.ebiom.2021.103678

Keywords

Human immunodeficiency virus type one; Viral eradication; Clustered regularly interspaced short palin-dromic repeat; RNA loaded Lipid nanoparticles (rLNP); Latent infection; CRISPR delivery

Funding

  1. University of Nebraska Foundation
  2. National Institutes of Health [T32 NS105594, 5R01MH121402, 1R01Al158160, R01 DA054535, PO1 DA028555, R01 NS126089, R01 NS36126, PO1 MH64570, P30 MH062261, 2R01 NS034239]
  3. National Institute for General Medical Science (NIGMS) [INBRE-P20GM103427-19]
  4. Fred & Pamela Buffett Cancer Center [P30 CA036727]

Ask authors/readers for more resources

The study developed a library of gRNAs designed to disrupt five HIV-1 exons, and validated their efficacy through various delivery methods. Results showed that multi-exon gRNA disruption delivered by LNPs enables effective excision of HIV-1.
Background: A barrier to HIV-1 cure rests in the persistence of proviral DNA in infected CD4+ leukocytes. The high HIV-1 mutation rate leads to viral diversity, immune evasion, and consequent antiretroviral drug resistance. While CRISPR-spCas9 can eliminate latent proviral DNA, its efficacy is limited by HIV strain diversity and precision target cell delivery. Methods: A library of guide RNAs (gRNAs) designed to disrupt five HIV-1 exons (omega t(1-2)/rev(1-2)/gp41) was constructed. The gRNAs were derived from a conseensus sequence of the transcriptional regulator tat from 4004 HIV-1 strains. Efficacy was affirmed by gRNA cell entry through transfection, electroporation, or by lentivirus or lipid nanoparticle (LNP) delivery. Treated cells were evaluated for viral excision by monitoring HIV-1 DNA, RNA, protein, and progeny virus levels. Findings: Virus was reduced in all transmitted founder strains by 82 and 94% after CRISPR TatDE transfection or lentivirus treatments, respectively. No recorded off-target cleavages were detected. Electroporation of TatDE ribonucleoprotein and delivery of LNP TatDE gRNA and spCas9 mRNA to latently infected cells resulted in up to 100% viral excision. Protection against HIV-1-challenge or induction of virus during latent infection, in primary or transformed CD4+ T cells or monocytes was achieved. We propose that multi-exon gRNA TatDE disruption delivered by LNPs enables translation for animal and human testing. Interpretation: These results provide proof of concept' for CRISPR gRNA treatments for HIV-1 elimination. The absence of full-length viral DNA by LNP delivery paired with undetectable off-target affirms the importance of payload delivery for effective viral gene editing. (C) 2021 The Author(s). Published by Elsevier B.V.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available