17
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      CRISPR-Cas9-mediated gene disruption of HIV-1 co-receptors confers broad resistance to infection in human T cells and humanized mice

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          In this preclinical study, we evaluated the efficacy and feasibility of creating broad human immunodeficiency virus (HIV) resistance by simultaneously disrupting the human CCR5 and CXCR4 genes, which encode cellular co-receptors required for HIV-1 infection. Using a clinically scalable system for transient ex vivo delivery of Cas9/guide RNA (gRNA) ribonucleoprotein (RNP) complexes, we demonstrated that CRISPR-mediated disruption of CCR5 and CXCR4 in T lymphocyte cells significantly reduced surface expression of the co-receptors, thereby establishing resistance to HIV-1 infection by CCR5 (R5)-tropic, CXCR4 (X4)-tropic, and dual (R5/X4)-tropic strains. Similarly, disruption of CCR5 alleles in human CD34 + hematopoietic stem and progenitor cells (HSPCs) successfully led to the differentiation of HIV-resistant macrophages. In a humanized mouse model under HIV-1 challenge, CXCR4-disrupted CD4 + T cells were enriched in the peripheral blood and spleen, indicating survival advantage because of resistance to viral infection. However, in human CD4 + T cells with both CCR5 and CXCR4 disruption, we observed poor engraftment in bone marrow, although significant changes were not observed in the lung, spleen, or peripheral blood. This study establishes a clinically scalable strategy for the dual knockout of HIV-1 co-receptors as a therapeutic strategy, while also raising caution of disrupting CXCR4, which may abate engraftment of CD4 + T cells in bone marrow.

          Graphical abstract

          Abstract

          CRISPR-mediated disruption of CCR5 and CXCR4 protects cells from infection of HIV-1 R5-trpoic, X4-tropic, and dual-tropic strains, but also abates engraftment of CD4 + T cells in bone marrow. This study establishes a clinically scalable strategy for the disruption of both HIV-1 co-receptors while also raising caution.

          Related collections

          Most cited references49

          • Record: found
          • Abstract: found
          • Article: not found

          Multiplex genome engineering using CRISPR/Cas systems.

          Functional elucidation of causal genetic variants and elements requires precise genome editing technologies. The type II prokaryotic CRISPR (clustered regularly interspaced short palindromic repeats)/Cas adaptive immune system has been shown to facilitate RNA-guided site-specific DNA cleavage. We engineered two different type II CRISPR/Cas systems and demonstrate that Cas9 nucleases can be directed by short RNAs to induce precise cleavage at endogenous genomic loci in human and mouse cells. Cas9 can also be converted into a nicking enzyme to facilitate homology-directed repair with minimal mutagenic activity. Lastly, multiple guide sequences can be encoded into a single CRISPR array to enable simultaneous editing of several sites within the mammalian genome, demonstrating easy programmability and wide applicability of the RNA-guided nuclease technology.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            DNA targeting specificity of RNA-guided Cas9 nucleases.

            The Streptococcus pyogenes Cas9 (SpCas9) nuclease can be efficiently targeted to genomic loci by means of single-guide RNAs (sgRNAs) to enable genome editing. Here, we characterize SpCas9 targeting specificity in human cells to inform the selection of target sites and avoid off-target effects. Our study evaluates >700 guide RNA variants and SpCas9-induced indel mutation levels at >100 predicted genomic off-target loci in 293T and 293FT cells. We find that SpCas9 tolerates mismatches between guide RNA and target DNA at different positions in a sequence-dependent manner, sensitive to the number, position and distribution of mismatches. We also show that SpCas9-mediated cleavage is unaffected by DNA methylation and that the dosage of SpCas9 and sgRNA can be titrated to minimize off-target modification. To facilitate mammalian genome engineering applications, we provide a web-based software tool to guide the selection and validation of target sequences as well as off-target analyses.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions

              Base editing is a recently developed approach to genome editing that uses a fusion protein containing a catalytically defective Streptococcus pyogenes Cas9, a cytidine deaminase, and an inhibitor of base excision repair to induce programmable, single-nucleotide changes in the DNA of living cells without generating double-strand DNA breaks, without requiring a donor DNA template, and without inducing an excess of stochastic insertions and deletions 1 . Here we report the development of five new C→T (or G→A) base editors that use natural and engineered Cas9 variants with different protospacer-adjacent motif (PAM) specificities to expand the number of sites that can be targeted by base editing by 2.5-fold. Additionally, we engineered new base editors containing mutated cytidine deaminase domains that narrow the width of the apparent editing window from approximately 5 nucleotides to as little as 1 to 2 nucleotides, enabling the discrimination of neighboring C nucleotides that would previously be edited with comparable efficiency, thereby doubling the number of disease-associated target Cs that can be corrected preferentially over nearby non-target Cs. Collectively, these developments substantially increase the targeting scope of base editing and establish the modular nature of base editors.
                Bookmark

                Author and article information

                Contributors
                Journal
                Mol Ther Methods Clin Dev
                Mol Ther Methods Clin Dev
                Molecular Therapy. Methods & Clinical Development
                American Society of Gene & Cell Therapy
                2329-0501
                22 January 2022
                10 March 2022
                22 January 2022
                : 24
                : 321-331
                Affiliations
                [1 ]Center for Gene Therapy, Beckman Research Institute of City of Hope, Duarte, CA, USA
                [2 ]Irell and Manella School of Biological Sciences, Duarte, CA, USA
                Author notes
                []Corresponding author John C. Burnett, Center for Gene Therapy, Beckman Research Institute of City of Hope, Duarte, CA, USA jburnett@ 123456coh.org
                Article
                S2329-0501(22)00013-4
                10.1016/j.omtm.2022.01.012
                8847835
                35229006
                f3ffd602-aa13-4861-b418-f33454980e41
                © 2022 The Authors

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 26 October 2021
                : 21 January 2022
                Categories
                Original Article

                hiv,crispr,ccr5,cxcr4,gene editing,bone marrow,macrophage,t cells,adoptive t cell therapy

                Comments

                Comment on this article