A simultaneous knockout knockin genome editingstrategy in HSPCs potently inhibits CCR5- andCXCR4-tropic HIV-1 infection

Authors:
Dudek AM, Feist WN, Sasu EJ, Luna SE, Ben-Efraim K, Bak RO, Cepika AM, Porteus MH
In:
Source: Cell Stem Cell
Publication Date: (2024)
Issue: 31(4): 499-518.e6
Research Area:
Cancer Research/Cell Biology
Immunotherapy / Hematology
Basic Research
Molecular Biology
Regenerative medicine
Cells used in publication:
CD34+ cell, human
Species: human
Tissue Origin: blood
Culture Media:
Platform:
4D-Nucleofector® X-Unit
Experiment

CD4+ T cell Isolation and Culture
CD4+ T cells were isolated from LRS chambers obtained from healthy donors at the Stanford Blood Center. Peripheral blood mononuclear cells (PBMCs) were isolated on a Ficoll density gradient followed by CD4+ cell enrichment through negative selection using magnetic separation with LS columns (Miltenyi) and cryopreservation in Bambanker cell freezing media (Wako Chemicals). T cells were thawed and cultured at 106 cells/mL in X-VIVO15 with gentamicin (Lonza), 5% human serum (Corning) and 100 IU mL–1 human IL-2 (Peprotech) with (HIV-optimized culture) or without (Standard culture) 40 ng/mL IL-7 and 10 ng/mL IL-12. At thaw, CD4+ T cells were activated with anti-CD3/anti-CD28 paramagnetic beads for standard culture conditions (Dynabeads Human T Cell Activator, Gibco) or on culture plates coated with Ultra-LEAF TM Purified anti-human CD3 (Biolegend #317326, clone OKT3, lot B329667) for HIV-optimized culture. Cells were cultured at 37°C with 5% CO2 and ambient O2, and replated at optimum cell density every 2-3 days with fresh complete media. For editing experiments, cells were activated in culture for three days prior to nucleofection, and activation beads were removed before electroporation where relevant.

CCR5 knockout
sgRNAs were acquired from TriLink Biotechnologies with 20-O-methyl-30-phosphorothioate modifications at the three terminal nucleotides of both ends and purified using HPLC. Guides sequences used for editing CCR5 were GCAGCATAGTGAGCCCAGAA (internal guide) or TGACATCAATTATTATACAT (start site guide). High-fidelity Cas9 protein was purchased from Aldeveron (SpyFi Cas9). RNPs were generated by precomplexing purified sgRNAs with HiFi SpCas9 protein at a molar ratio of 2.5:1 for 15-30 minutes at room temperature. RNPs were electroporated at a Cas9 concentration of 300 µg per mL using a 4D Nucleofector (Lonza) in 16-cuvette strips. 250,000 HSPCs or 106 activated T cells were nucleofected in P3 solution with program DZ100 or EO-115, respectively, with or without RNP and rAAV6 as noted.

Abstract

Allogeneic hematopoietic stem and progenitor cell transplant (HSCT) of CCR5 null (CCR5D32) cells can be curative for HIV-1-infected patients. However, because allogeneic HSCT poses significant risk, CCR5D32 matched bone marrow donors are rare, and CCR5D32 transplant does not confer resistance to the CXCR4-tropic virus, it is not a viable option for most patients. We describe a targeted Cas9/AAV6-based genome editing strategy for autologous HSCT resulting in both CCR5- and CXCR4-tropic HIV-1 resistance. Edited human hematopoietic stem and progenitor cells (HSPCs) maintain multi-lineage repopulation capacity in vivo, and edited primary human T cells potently inhibit infection by both CCR5-tropic and CXCR4-tropic HIV-1. Modification rates facilitated complete loss of CCR5-tropic replication and up to a 2,000-fold decrease in CXCR4-tropic replication without CXCR4 locus disruption. This multi-factor editing strategy in HSPCs could provide a broad approach for autologous HSCT as a functional cure for both CCR5-tropic and CXCR4-tropic HIV-1 infections.