scholarly journals Human microglia extensively reconstitute in humanized BLT mice with human interleukin-34 transgene and support HIV-1 brain infection

2021 ◽  
Author(s):  
Jianshui Zhang ◽  
Saroj Chandra Lohani ◽  
Yilun Cheng ◽  
Tao Wang ◽  
Lili Guo ◽  
...  

AbstractHumanized bone marrow-liver-thymic (hu-BLT) mice develop a functional immune system in periphery but have a limited reconstitution of human myeloid cells, especially microglia, in CNS. Further, whether bone marrow derived hematopoietic stem and progenitor cells (HSPCs) can enter the brain and differentiate into microglia in adults remains controversial. To close these gaps, in this study we unambiguously demonstrated that human microglia in CNS were extensively reconstituted in adult NOG mice with human interleukin-34 transgene (hIL34 Tg) from circulating CD34+ HSPCs but no in hu-BLT NOG mice, providing strong evidence that human CD34+ HSPCs can enter adult brain and differentiate into microglia in CNS in the presence of hIL34. Further, the human microglia in the CNS of hu-BLT-hIL34 NOG mice robustly supported HIV-1 infection reenforcing the notion that microglia are the most important target cells of HIV-1 in CNS and demonstrating its great potential as an in vivo model for studying HIV-1 pathogenesis and evaluating curative therapeutics in both periphery and CNS compartments.

2021 ◽  
Vol 12 ◽  
Author(s):  
Jianshui Zhang ◽  
Saroj Chandra Lohani ◽  
Yilun Cheng ◽  
Tao Wang ◽  
Lili Guo ◽  
...  

Humanized bone marrow-liver-thymic (hu-BLT) mice develop a functional immune system in periphery, nevertheless, have a limited reconstitution of human myeloid cells, especially microglia, in CNS. Further, whether bone marrow derived hematopoietic stem and progenitor cells (HSPCs) can enter the brain and differentiate into microglia in adults remains controversial. To close these gaps, in this study we unambiguously demonstrated that human microglia in CNS were extensively reconstituted in adult NOG mice with human interleukin-34 transgene (hIL34 Tg) from circulating CD34+ HSPCs, nonetheless not in hu-BLT NOG mice, providing strong evidence that human CD34+ HSPCs can enter adult brain and differentiate into microglia in CNS in the presence of hIL34. Further, the human microglia in the CNS of hu-BLT-hIL34 NOG mice robustly supported HIV-1 infection reenforcing the notion that microglia are the most important target cells of HIV-1 in CNS and demonstrating its great potential as an in vivo model for studying HIV-1 pathogenesis and evaluating curative therapeutics in both periphery and CNS compartments.


2015 ◽  
Vol 89 (13) ◽  
pp. 6761-6772 ◽  
Author(s):  
Renier Myburgh ◽  
Sandra Ivic ◽  
Michael S. Pepper ◽  
Gustavo Gers-Huber ◽  
Duo Li ◽  
...  

ABSTRACTGene-engineered CD34+hematopoietic stem and progenitor cells (HSPCs) can be used to generate an HIV-1-resistant immune system. However, a certain threshold of transduced HSPCs might be required for transplantation into mice for creating an HIV-resistant immune system. In this study, we combined CCR5 knockdown by a highly efficient microRNA (miRNA) lentivector with pretransplantation selection of transduced HSPCs to obtain a rather pure population of gene engineered CD34+cells. Low-level transduction of HSPCs and subsequent sorting by flow cytometry yielded >70% transduced cells. Mice transplanted with these cells showed functional and persistent resistance to a CCR5-tropic HIV strain: viral load was significantly decreased over months, and human CD4+T cells were preserved. In one mouse, viral mutations, resulting presumably in a CXCR4-tropic strain, overcame HIV resistance. Our results suggest that HSPC-based CCR5 knockdown may lead to efficient control of HIVin vivo. We overcame a major limitation of previous HIV gene therapy in humanized mice in which only a proportion of the cells in chimeric micein vivoare anti-HIV engineered. Our strategy underlines the promising future of gene engineering HIV-resistant CD34+cells that produce a constant supply of HIV-resistant progeny.IMPORTANCEMajor issues in experimental long-termin vivoHIV gene therapy have been (i) low efficacy of cell transduction at the time of transplantation and (ii) transduction resulting in multiple copies of heterologous DNA in target cells. In this study, we demonstrated the efficacy of a transplantation approach with a selection step for transduced cells that allows transplantation of an enriched population of HSPCs expressing a single (low) copy of a CCR5 miRNA. Efficient maintenance of CD4+T cells and a low viral titer resulted only when at least 70% of the HIV target cells were genetically modified. These findings imply that clinical protocols of HIV gene therapy require a selective enrichment of genetically targeted cells because positive selection of modified cells is likely to be insufficient below this threshold. This selection approach may be beneficial not only for HIV patients but also for other patients requiring transplantation of genetically modified cells.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1758-1758
Author(s):  
Axel Schambach ◽  
Bernhard Schiedlmeier ◽  
Jens Bohne ◽  
Dorothee von Laer ◽  
Geoff Margison ◽  
...  

Abstract T20 is a 36-amino-acid peptide that binds to HIV-1 gp41 and thereby acts as a fusion inhibitor, thus mediating potent and selective inhibition of HIV-1 entry in vitro and in vivo. An extended peptide expressed as an artificial, membrane-bound molecule (mbC46) efficiently inhibits HIV infection of primary human T-cells following retroviral vector mediated gene transfer (Egelhofer et al., J Virol, 2004). To develop an even more stringent approach to HIV gene therapy, we targeted hematopoietic stem cells. In 3 experimental groups of C57BL/6 mice (9 animals/group), we investigated the long-term toxicity of murine bone marrow cells transduced with M87o, a therapeutic vector designed to coexpress mbC46 and an HIV-derived RNA RRE-decoy to inhibit HIV replication. As controls we used the same vector containing an inactive C46 peptide and mock-transduced cells. Blood samples were collected monthly. Donor chimerism and transgene expression in multiple lineages were determined by FACS analysis and transgene integration was measured by real time PCR. Six months after transplantation, 4 mice per group were sacrificed and the remaining 5 mice per group were observed for another 6 months. In addition to the parameters mentioned above, we performed complete histopathology, blood counts and clinical biochemistry. Donor chimerism in all groups ranged from 82 – 94% (day 190 and day 349). In the M87o group, 60% of donor cells expressed mbC46. FACS data showed persisting transgene expression in T-cells (CD4, CD8, 65%), B-cells (B220, 46%), myeloid cells (CD11b, 68%), platelets (CD41, 19%), and RBC (60%) of the peripheral blood and bone marrow cells. Highly sustained gene marking (2–4 copies/genome) was noticed on day 190. To reveal latent malignant clones potentially originating from side effects of the genetic manipulation, 1x106 bone marrow cells from 4 primary recipients were transplanted into lethally irradiated secondary recipients (3 recipients/primary mouse) and these mice were observed for 8 months. All together, we could not observe any evidence for leukemogenic capacity. Analysis of peripheral blood and bone marrow showed a similar transgene expression pattern compared to the primary mice. To generate a complete chimerism of transgenic cells, we chose the human drug resistance gene methylguanine-methyltransferase (MGMT, P140K) to select for mbC46-transduced stem cells in vitro and in vivo. Different coexpression strategies were tested. Function of the MGMT protein was confirmed in a quantitative alkyltransferase assay and in a cytotoxicity assay using BCNU or temozolomide. In vitro selection of transduced 32D and PM1 cells with benzylguanine and BCNU showed >95% positive cells with evidence of polyclonal survival. Transduced PM1 cells underwent an HIV challenge assay. In vivo experiments in a murine bone marrow transplantation setting are ongoing to determine the potency and safety of combined retroviral expression of mbC46 and MGMT in relevant preclinical models. Successful conclusion of these studies will hopefully result in a phase I clinical trial testing the concept of generating an HIV-resistant autologous hematopoiesis.


eLife ◽  
2019 ◽  
Vol 8 ◽  
Author(s):  
Mark S Ladinsky ◽  
Wannisa Khamaikawin ◽  
Yujin Jung ◽  
Samantha Lin ◽  
Jennifer Lam ◽  
...  

Immune progenitor cells differentiate in bone marrow (BM) and then migrate to tissues. HIV-1 infects multiple BM cell types, but virus dissemination within BM has been poorly understood. We used light microscopy and electron tomography to elucidate mechanisms of HIV-1 dissemination within BM of HIV-1–infected BM/liver/thymus (BLT) mice. Tissue clearing combined with confocal and light sheet fluorescence microscopy revealed distinct populations of HIV-1 p24-producing cells in BM early after infection, and quantification of these populations identified macrophages as the principal subset of virus-producing cells in BM over time. Electron tomography demonstrated three modes of HIV-1 dissemination in BM: (i) semi-synchronous budding from T-cell and macrophage membranes, (ii) mature virus association with virus-producing T-cell uropods contacting putative target cells, and (iii) macrophages engulfing HIV-1–producing T-cells and producing virus within enclosed intracellular compartments that fused to invaginations with access to the extracellular space. These results illustrate mechanisms by which the specialized environment of the BM can promote virus spread locally and to distant lymphoid tissues.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1804-1804
Author(s):  
Ghanwa Khawaja ◽  
Yang Jo Chung ◽  
Eunsil Park ◽  
Micheal Difilippantonio ◽  
James H. Doroshow ◽  
...  

Abstract The myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis, peripheral blood cytopenias, dysplasia and a propensity for transformation to acute myeloid leukemia (AML). MDS is frequently associated with epigenetic gene silencing via methylation of cytosine residues in gene regulatory regions, and DNA methyl-transferase 1 (DNMT1) inhibitors, such as 5'azacytidine and 5-aza-2'-deoxycytidine (decitabine, DAC), are two of the three agents that are FDA approved for treatment of MDS. Although these drugs are not curative, they induce hematological improvement or improved survival in a significant fraction of MDS patients. Two novel, thiol-substituted 2'-deoxycytidine (dCyd) analogs designated T-dCyd (4'-thio-2'-deoxycytidine) and Aza-T-dCyd (5-aza-4'-thio-2'-deoxycytidine) were synthesized and shown to be potent DNMT1 inhibitors in vitro. We evaluated these drugs in vivo using the NUP98-HOXD13 (NHD13) mouse model for MDS. To mimic human MDS hematopoiesis, in which a portion of the hematopoietic output is provided by the MDS clone, and a portion provided by normal, non-MDS cells, we transplanted wild-type (WT) mice with a mixture of WT murine hematopoietic cells and NHD13 (MDS) hematopoietic cells. This bone marrow transplant (BMT) produces chimaeric recipients with bone marrow comprised of hematopoietic cells derived from both the MDS clone as well as normal hematopoietic precursors. WT and MDS cells in the mice can be distinguished by differential CD45 alleles (CD45.1 and CD45.2, respectively), which enables analysis and purification of the MDS and WT cells; this feat is not easily achieved with human MDS patient samples, which lack cell surface antigens specific for the MDS clone. At 8 weeks post-transplant; engraftment of MDS cells was documented by the presence of CD45.2+ cells in the peripheral blood, and the starting CBCs showed signs consistent with MDS including peripheral blood cytopenia and macrocytosis. Mice were randomly assigned to one of the three groups. 1) PBS, 2) T-dCyd, 3) Aza-T-dCyd. T-Cyd was dosed at 4 mg/kg/d intraperitoneally (IP) on weekdays for 2 weeks (10 doses), followed by three weeks rest; this constituted one cycle of therapy. Aza-T-dCyd was administered on the same schedule at 4 mg/kg/d IP. Flow cytometry and CBC were assessed on day 21 of each cycle, and treatment continued for up to one year, or until mice were humanely euthanized due to tachypnea, lethargy, or other signs of AML. Between four and six mice were treated per group, and the entire experiment was repeated three times and results pooled for T-dCyd, once for Aza-T-dCyd. The T-dCyd treated chimaeric mice showed significantly enhanced overall survival associated with hematological improvement including hemoglobin concentration, platelet and absolute neutrophil count compared to PBS treated mice (median survival 45.4 vs 28 weeks, p=0.0187). In addition to a survival advantage, AML onset was significantly delayed in the T-dCyd treated mice (median time to AML transformation 35 weeks for PBS vs unreached for T-dCyd, p=0.0111), although there was no significant change in MDS (CD45.2) engraftment between the T-dCyd and PBS treated mice. For Aza-T-dCyd group, we did not detect a survival benefit nor hematologic improvement, although we suspect this may have been secondary to unexpected toxicity at the selected dose. In sum, these results demonstrate the utility of chimaeric WT/MDS mice as a pre-clinical model for human MDS, and show that treatment with T-dCyd, a new DNMT1 inhibitor, leads to a survival advantage, hematologic improvement, and delayed transformation to AML. Disclosures Aplan: NIH Office of Technolgy Transfer: Employment, Patents & Royalties: NUP98-HOXD13 mice.


Author(s):  
Oliver J. Herd ◽  
Gulab Fatima Rani ◽  
James P Hewitson ◽  
Karen Hogg ◽  
Andrew P Stone ◽  
...  

Immune thrombocytopenia (ITP) is an acquired autoimmune condition characterized by both reduced platelet production and the destruction of functionally normal platelets by sustained attack from the immune system. However, the effect of prolonged ITP on the more immature hematopoietic progenitors remains an open area of investigation. Using a murine in vivo model of extended ITP, we reveal that ITP progression drives considerable progenitor expansion and bone marrow (BM) remodelling. Single cell assays using Lin-Sca1+c-Kit+CD48-CD150+ long-term hematopoietic stem cells (LT-HSCs) revealed elevated LT-HSC activation and proliferation in vitro. However, the increased activation did not come at the expense of LT-HSC functionality as measured by in vivo serial transplantations. ITP progression was associated with considerable BM vasodilation and angiogenesis, as well as a 2-fold increase in local production of CXCL12; a cytokine essential for LT-HSC function and BM homing expressed at high levels by LepR+ BM stromal cells. This was associated with a 1.5-fold increase in LepR+ BM stromal cells and a 5.5-fold improvement in progenitor homing to the BM. Whereas the increase in stromal cells was transient and reverted back to baseline after platelet count returned to normal, vasculature changes in the BM persisted. Together, these studies demonstrate that LT-HSCs expand in response to ITP, and that LT-HSC functionality during sustained hematopoietic stress is maintained through an adapting BM microenvironment.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 238-238
Author(s):  
Frederic Barabe ◽  
James A. Kennedy ◽  
John E. Dick

Abstract Identification of genes and translocations involved in human leukemia, as well as classification and clustering by gene arrays, have greatly evolved in the past years. However, the mechanisms of human leukemogenesis remain to be elucidated and the failure to develop an in vivo model where primary human hematopoietic cells are transformed into leukemic cells represents a significant limitation. Using a retrovirus encoding the oncogene MLL-ENL resulting from the t(11;19)(q23;p13.3) translocation found in acute myeloid leukemias (AML) as well as in acute lymphoblastic leukemias (ALL) of B or T cell origin, we infected lineage-negative cord blood cells and injected those cells into sub-lethally irradiated NOD/SCID mice. 15 to 20 weeks after injection, all the mice developed an aggressive pro-B acute lymphoblastic leukemia characterized by immature B cells (CD10+, CD19+, CD20−, IgD−, IgM−) involving more than 90% of bone marrow. Spleen and thymus were increased in size and infiltrated with >90% leukemic cells. Furthermore, analysis of the lungs and liver showed significant infiltration of these organs. Transplantation of leukemic cells from primary mice to secondary recipients was able to recapitulate the disease with the same phenotype and the same organ involvement in a shorter period of time. If MLL-ENL transduced cells are grown in suspension culture with IL-3 and SCF, there is massive proliferation of cells blocked in differentiation along the monocytic lineage. In contrast to untransduced cells, colony-forming progenitors were maintained long term in these cultures and could be serially replated, suggestive of an enhanced capacity for self-renewal. After 50 to 70 days in culture, these cells were injected in NOD/SCID mice and mice were analyzed after 12 to 15 weeks. Monoblastic cells were engrafted in the bone marrow and spleen with the same phenotype of the cultured cells (CD33+, CD11b+, CD15+, HLA DR+). These cells were able to engraft secondary and tertiary recipients formally demostrating increased self-renewal capacity of the transformed stem cell. In a limited number of primary mice, transplanted with high cell doses, AML developed at 15 weeks post-transplant. To our knowledge, these results provide the first in vivo model where human hematopietic stem/progenitor cells are transformed into leukemia. Remarkably, depending on the cellular environment, MLL-ENL can induce ALL or AML in primary cells as a sole genetic event, although we cannot rule out the spontaneous acquistion of additional co-operating genetic or epigenetic abnormalities. This model provides a significant step forward to understand the mechanisms involved in human leukemogenesis.


Blood ◽  
1996 ◽  
Vol 87 (10) ◽  
pp. 4136-4142 ◽  
Author(s):  
I Kawashima ◽  
ED Zanjani ◽  
G Almaida-Porada ◽  
AW Flake ◽  
H Zeng ◽  
...  

Using in utero transplantation into fetal sheep, we examined the capability of human bone marrow CD34+ cells fractionated based on Kit protein expression to provide long-term in vivo engraftment. Twelve hundred to 5,000 CD34+ Kit-, CD34+ Kit(low), and CD34+ Kit(high) cells were injected into a total of 14 preimmune fetal sheep recipients using the amniotic bubble technique. Six fetuses were killed in utero 1.5 months after bone marrow cell transplantation. Two fetuses receiving CD34+ Kit(low) cells showed signs of engraftment according to analysis of CD45+ cells in their bone marrow cells and karyotype studies of the colonies grown in methylcellulose culture. In contrast, two fetuses receiving CD34+ Kit(high) cells and two fetuses receiving CD34+ Kit- cells failed to show evidence of significant engraftment. Two fetuses were absorbed. A total of six fetuses receiving different cell populations were allowed to proceed to term, and the newborn sheep were serially examined for the presence of chimerism. Again, only the two sheep receiving CD34+ Kit(low) cells exhibited signs of engraftment upon serial examination. Earlier in studies of murine hematopoiesis, we have shown stage-specific changes in Kit expression by the progenitors. The studies of human cells reported here are in agreement with observations in mice, and indicate that human hematopoietic stem cells are enriched in the Kit(low) population.


Blood ◽  
1997 ◽  
Vol 89 (11) ◽  
pp. 3919-3924 ◽  
Author(s):  
Jean C.Y. Wang ◽  
Monica Doedens ◽  
John E. Dick

Abstract We have previously reported the development of in vivo functional assays for primitive human hematopoietic cells based on their ability to repopulate the bone marrow (BM) of severe combined immunodeficient (SCID) and nonobese diabetic/SCID (NOD/SCID) mice following intravenous transplantation. Accumulated data from gene marking and cell purification experiments indicate that the engrafting cells (defined as SCID-repopulating cells or SRC) are biologically distinct from and more primitive than most cells that can be assayed in vitro. Here we demonstrate through limiting dilution analysis that the NOD/SCID xenotransplant model provides a quantitative assay for SRC. Using this assay, the frequency of SRC in cord blood (CB) was found to be 1 in 9.3 × 105 cells. This was significantly higher than the frequency of 1 SRC in 3.0 × 106 adult BM cells or 1 in 6.0 × 106 mobilized peripheral blood (PB) cells from normal donors. Mice transplanted with limiting numbers of SRC were engrafted with both lymphoid and multilineage myeloid human cells. This functional assay is currently the only available method for quantitative analysis of human hematopoietic cells with repopulating capacity. Both CB and mobilized PB are increasingly being used as alternative sources of hematopoietic stem cells in allogeneic transplantation. Thus, the findings reported here will have important clinical as well as biologic implications.


Sign in / Sign up

Export Citation Format

Share Document