14 research outputs found

    Engineering HIV-1-resistant T-cells from short-hairpin RNA-expressing hematopoietic stem/progenitor cells in humanized BLT mice

    Get PDF
    Down-regulation of the HIV-1 coreceptor CCR5 holds significant potential for long-term protection against HIV-1 in patients. Using the humanized bone marrow/liver/thymus (hu-BLT) mouse model which allows investigation of human hematopoietic stem/progenitor cell (HSPC) transplant and immune system reconstitution as well as HIV-1 infection, we previously demonstrated stable inhibition of CCR5 expression in systemic lymphoid tissues via transplantation of HSPCs genetically modified by lentiviral vector transduction to express short hairpin RNA (shRNA). However, CCR5 down-regulation will not be effective against existing CXCR4-tropic HIV-1 and emergence of resistant viral strains. As such, combination approaches targeting additional steps in the virus lifecycle are required. We screened a panel of previously published shRNAs targeting highly conserved regions and identified a potent shRNA targeting the R-region of the HIV-1 long terminal repeat (LTR). Here, we report that human CD4+ T-cells derived from transplanted HSPC engineered to co-express shRNAs targeting CCR5 and HIV-1 LTR are resistant to CCR5- and CXCR4- tropic HIV-1-mediated depletion in vivo. Transduction with the combination vector suppressed CXCR4- and CCR5- tropic viral replication in cell lines and peripheral blood mononuclear cells in vitro. No obvious cytotoxicity or interferon response was observed. Transplantation of combination vector-transduced HSPC into hu-BLT mice resulted in efficient engraftment and subsequent stable gene marking and CCR5 down-regulation in human CD4+ T-cells within peripheral blood and systemic lymphoid tissues, including gut-associated lymphoid tissue, a major site of robust viral replication, for over twelve weeks. CXCR4- and CCR5- tropic HIV-1 infection was effectively inhibited in hu-BLT mouse spleen-derived human CD4+ T-cells ex vivo. Furthermore, levels of gene-marked CD4+ T-cells in peripheral blood increased despite systemic infection with either CXCR4- or CCR5- tropic HIV-1 in vivo. These results demonstrate that transplantation of HSPCs engineered with our combination shRNA vector may be a potential therapy against HIV disease.This work was supported by grants from the California Institute for Regenerative Medicine (CIRM grant DR1-01431 to ISYC), the National Institutes of Health (1RO1HL086409 and 3RO1HL086409-03S1 to DSA and 5T32AI060567), and the University of California Los Angeles AIDS Institute/Center for AIDS Research (5P30AI028697). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript

    Ectopic expression of anti-HIV-1 shRNAs protects CD8 + T cells modified with CD4ζ CAR from HIV-1 infection and alleviates impairment of cell proliferation

    No full text
    Chimeric antigen receptors (CARs) are artificially engineered receptors that confer a desired specificity to immune effector T cells. As an HIV-1-specific CAR, CD4ζ CAR has been extensively tested in vitro as well as in clinical trials. T cells modified with this CAR mediated highly potent anti-HIV-1 activities in vitro and were well-tolerated in vivo, but exerted limited effects on viral load and reservoir size due to poor survival and/or functionality of the transduced cells in patients. We hypothesize that ectopic expression of CD4ζ on CD8(+) T cells renders them susceptible to HIV-1 infection, resulting in poor survival of those cells. To test this possibility, highly purified CD8(+) T cells were genetically modified with a CD4ζ-encoding lentiviral vector and infected with HIV-1. CD8(+) T cells were vulnerable to HIV-1 infection upon expression of CD4ζ as evidenced by elevated levels of p24(Gag) in cells and culture supernatants. Concurrently, the number of CD4ζ-modified CD8(+) T cells was reduced relative to control cells upon HIV-1 infection. To protect these cells from HIV-1 infection, we co-expressed two anti-HIV-1 shRNAs previously developed by our group together with CD4ζ. This combination vector was able to suppress HIV-1 infection without impairing HIV-1-dependent effector activities of CD4ζ. In addition, the number of CD4ζ-modified CD8(+) T cells maintained similar levels to that of the control even under HIV-1 infection. These results suggest that protecting CD4ζ-modified CD8(+) T cells from HIV-1 infection is required for prolonged HIV-1-specific immune surveillance

    Down-regulation of CCR5 and inhibition of HIV-1 replication in MOLT4-CCR5 cells and PBMCs by Dual sh1005/sh516.

    No full text
    <p><b>A.</b> MOLT4/CCR5 (1×10<sup>5</sup>) cells were transduced with lentiviral vectors at MOI 0.5. EGFP and CCR5 expression was assessed three days post-transduction. CCR5 MFIs shown below plots as “MFI: MFI<sub>EGFP−</sub>/MFI<sub>EGFP+</sub>.” Representative data of three independent experiments. <b>B.</b> Sorted EGFP<sup>+</sup> cells (2×10<sup>5</sup>) were infected with either HIV-1<sub>NL4-3</sub> at MOI 0.5 and HIV-1<sub>NFNSX SL9</sub> at MOI 5.0. Levels of p24 antigen in culture supernatants were measured by ELISA four and seven days post-infection. Errors bars: mean + SD. <b>C.</b> IL-2/PHA stimulated PBMCs (4×10<sup>5</sup>) were transduced with lentiviral vectors at MOI 0.6–1.0. EGFP and CCR5 expression was measured at seven days post-transduction. CCR5 MFIs shown as in <b>A</b>. Representative data showing CCR5 expression in vector-transduced PBMCs. <b>D.</b> Sorted EGFP<sup>+</sup> cells (5×10<sup>4</sup>) were infected with either HIV-1<sub>NFNSX SL9</sub> at MOI 5.0, HIV-1<sub>JR-CSF</sub> at MOI 1.0, or HIV-1<sub>NL4-3</sub> at MOI 0.1. p24 production was measured as in <b>B</b>.</p

    Vector stability and the effects of sh1005/sh516 co-expression on cell viability and HSPC differentiation potential.

    No full text
    <p>IL-2/PHA-stimulated PBMCs (4×10<sup>5</sup>) were transduced with lentiviral vectors at MOI 0.6–1.0. <b>A.</b> Representative data showing EGFP expression in vector-transduced PBMCs over time. <b>B.</b> Vector-transduced cells were subjected to CytoTox-Glo<sup>TM</sup> Cytotoxicity Assay (Promega) four and six days post-transduction. Relative cytotoxicities were calculated by dividing the dead cell count by the total cell count and normalizing to that of mock-transduced cells. 1 μM Staurosporine (STS) and U6 promoter-driven sh1005 expression (U6-sh1005) served as positive controls. Error bars: mean + SD. <b>C.</b> OAS1 mRNA expression relative to β-actin was assessed by qRT-PCR analysis of total RNA isolated from vector-transduced PBMCs. PBMCs harvested two days post-electroporation with 500 pg/μL poly(I:C) served as positive control. Values were normalized to those of mock-transduced cells. Error bars: mean + SD. <b>D.</b> Cytokine-pre-stimulated mPB-CD34<sup>+</sup> cells were transduced with lentiviral vectors at MOI 10. HSPC differentiation potential was assessed by counting colony forming units produced from mock- and vector- transduced mPB-CD34<sup>+</sup> cells plated on semi-solid methylcellulose plates one day after transduction. CFU-GEMM: granulocyte/erythrocyte/macrophage/megakaryocyte colony forming units. C/BFU-E: erythroid colony/burst forming units. CFU-GM: granulocyte/monocyte colony forming units. Error bars: range of values between duplicate samples. Representative data of three independent experiments.</p

    Down-regulation of CCR5 in systemic lymphoid tissues and inhibition of HIV-1 replication <i>ex vivo</i> by Dual sh1005/sh516.

    No full text
    <p>Tissues were isolated from a Dual sh1005/sh516-transduced HSPC-transplanted mouse at eleven weeks post-transplantation. <b>A.</b> Representative data showing CCR5 expression in EGFP- and mCherry- marked human CD4<sup>+</sup>CD3<sup>+</sup>CD45<sup>+</sup>CD19<sup>−</sup> T-cells within a gated lymphocyte population. <b>B.</b> Splenocytes were depleted of human CD8<sup>+</sup> cells and murine CD45<sup>+</sup> cells and then stimulated with PHA/IL-2 for two days. Five days later, EGFP<sup>+</sup> and mCherry<sup>+</sup> cells were then sorted by FACS at >97% purities. Sorted cells (5×10<sup>4</sup>) were then infected with either HIV-1<sub>NL4-3</sub> at MOI 0.5, HIV-1<sub>NFNSX SL9</sub> at MOI 5.0, or HIV-1<sub>JR-CSF</sub> at MOI 1.0 for four hours. Cells were then washed five times before culturing. HIV-1 replication was monitored by p24 ELISA analysis of culture supernatants at four and seven days post-infection. Error bars: mean + SD. Representative data of three independent experiments.</p

    Reconstitution of Dual sh1005/sh516-transduced HSPCs in humanized BLT mice.

    No full text
    <p><b>A.</b> Schematic of generating vector-transduced HSPC-transplanted hu-BLT mouse. NSG mouse is treated with Busulfan 24 hours pre-transplantation. CD34<sup>+</sup> and CD34<sup>−</sup> cells are isolated from human fetal liver (FL). CD34<sup>+</sup> cells are transduced with either therapeutic (EGFP-marked) or control (mCherry-marked) vectors. Therapeutic vector- and control vector- transduced CD34<sup>+</sup> cells are mixed at a 50 50 ratio. The cell mixture is then 1) combined with CD34<sup>−</sup> cells, solidified with Matrigel, and implanted under the kidney capsule with a human fetal thymus segment (FT) and also 2) intravenously injected. <b>B.</b> EGFP and mCherry reporter gene expression was monitored in human CD45<sup>+</sup>, CD3<sup>+</sup>CD45<sup>+</sup>, and CD19<sup>+</sup>CD45<sup>+</sup> cells within a gated lymphocyte population in peripheral blood at twelve weeks post-transplantation. CD4/CD8 ratios were analyzed in mCherry- and EGFP- marked CD3<sup>+</sup>CD45<sup>+</sup> cells. Data were generated from n = 13 mice for both Mono sh1005- and Dual sh1005/sh516- HSPC-transplanted animals from an aggregate of three donors. Error bars: mean + standard error of mean (SEM) in n = 13 per group.</p
    corecore