Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Publication year range
1.
Cell ; 153(5): 1134-48, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23664764

ABSTRACT

Epigenetic mechanisms have been proposed to play crucial roles in mammalian development, but their precise functions are only partially understood. To investigate epigenetic regulation of embryonic development, we differentiated human embryonic stem cells into mesendoderm, neural progenitor cells, trophoblast-like cells, and mesenchymal stem cells and systematically characterized DNA methylation, chromatin modifications, and the transcriptome in each lineage. We found that promoters that are active in early developmental stages tend to be CG rich and mainly engage H3K27me3 upon silencing in nonexpressing lineages. By contrast, promoters for genes expressed preferentially at later stages are often CG poor and primarily employ DNA methylation upon repression. Interestingly, the early developmental regulatory genes are often located in large genomic domains that are generally devoid of DNA methylation in most lineages, which we termed DNA methylation valleys (DMVs). Our results suggest that distinct epigenetic mechanisms regulate early and late stages of ES cell differentiation.


Subject(s)
DNA Methylation , Embryonic Stem Cells/metabolism , Epigenomics , Gene Expression Regulation, Developmental , Animals , Cell Differentiation , Chromatin/metabolism , CpG Islands , Embryonic Stem Cells/cytology , Histones/metabolism , Humans , Methylation , Neoplasms/genetics , Promoter Regions, Genetic , Zebrafish/embryology
2.
Emerg Infect Dis ; 30(4): 721-731, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38526136

ABSTRACT

Genetically diverse simian arteriviruses (simarteriviruses) naturally infect geographically and phylogenetically diverse monkeys, and cross-species transmission and emergence are of considerable concern. Characterization of most simarteriviruses beyond sequence analysis has not been possible because the viruses fail to propagate in the laboratory. We attempted to isolate 4 simarteriviruses, Kibale red colobus virus 1, Pebjah virus, simian hemorrhagic fever virus, and Southwest baboon virus 1, by inoculating an immortalized grivet cell line (known to replicate simian hemorrhagic fever virus), primary macaque cells, macrophages derived from macaque induced pluripotent stem cells, and mice engrafted with macaque CD34+-enriched hematopoietic stem cells. The combined effort resulted in successful virus isolation; however, no single approach was successful for all 4 simarteriviruses. We describe several approaches that might be used to isolate additional simarteriviruses for phenotypic characterization. Our results will expedite laboratory studies of simarteriviruses to elucidate virus-host interactions, assess zoonotic risk, and develop medical countermeasures.


Subject(s)
Arterivirus , Animals , Mice , Arterivirus/genetics , Macaca , Macrophages , Cell Line
3.
Retrovirology ; 19(1): 17, 2022 08 10.
Article in English | MEDLINE | ID: mdl-35948929

ABSTRACT

Nonhuman primates (NHPs) are well-established basic and translational research models for human immunodeficiency virus (HIV) infections and pathophysiology, hematopoietic stem cell (HSC) transplantation, and assisted reproductive technologies. Recent advances in CRISPR/Cas9 gene editing technologies present opportunities to refine NHP HIV models for investigating genetic factors that affect HIV replication and designing cellular therapies that exploit genetic barriers to HIV infections, including engineering mutations into CCR5 and conferring resistance to HIV/simian immunodeficiency virus (SIV) infections. In this report, we provide an overview of recent advances and challenges in gene editing NHP embryos and discuss the value of genetically engineered animal models for developing novel stem cell-based therapies for curing HIV.


Subject(s)
Gene Editing , HIV Infections , Animals , CRISPR-Cas Systems , Humans , Primates , Stem Cells
4.
Blood ; 134(16): 1298-1311, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31416800

ABSTRACT

Therapeutic gene delivery to hematopoietic stem cells (HSCs) holds great potential as a life-saving treatment of monogenic, oncologic, and infectious diseases. However, clinical gene therapy is severely limited by intrinsic HSC resistance to modification with lentiviral vectors (LVs), thus requiring high doses or repeat LV administration to achieve therapeutic gene correction. Here we show that temporary coapplication of the cyclic resveratrol trimer caraphenol A enhances LV gene delivery efficiency to human and nonhuman primate hematopoietic stem and progenitor cells with integrating and nonintegrating LVs. Although significant ex vivo, this effect was most dramatically observed in human lineages derived from HSCs transplanted into immunodeficient mice. We further show that caraphenol A relieves restriction of LV transduction by altering the levels of interferon-induced transmembrane (IFITM) proteins IFITM2 and IFITM3 and their association with late endosomes, thus augmenting LV core endosomal escape. Caraphenol A-mediated IFITM downregulation did not alter the LV integration pattern or bias lineage differentiation. Taken together, these findings compellingly demonstrate that the pharmacologic modification of intrinsic immune restriction factors is a promising and nontoxic approach for improving LV-mediated gene therapy.


Subject(s)
Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/virology , Membrane Proteins/drug effects , Resveratrol/pharmacology , Transduction, Genetic/methods , Animals , Endosomes/drug effects , Endosomes/metabolism , Genetic Vectors , Heterografts , Humans , Lentivirus , Membrane Proteins/metabolism , Mice , Protein Transport/drug effects
5.
J Immunol ; 202(3): 770-776, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30578305

ABSTRACT

Human pluripotent stem cells (hPSCs) offer the potential to serve as a versatile and scalable source of T cells for immunotherapies, which could be coupled with genetic engineering technologies to meet specific clinical needs. To improve T cell production from hPSCs, it is essential to identify cell subsets that are highly enriched in T cell progenitors and those stages of development at which NOTCH activation induces the most potent T cells. In this study, we evaluated the efficacy of T cell production from cell populations isolated at different stages of hematopoietic differentiation, including mesoderm, hemogenic endothelium (HE), and multipotent hematopoietic progenitors. We demonstrate that KDRhiCD31- hematovascular mesodermal progenitors (HVMPs) with definitive hematopoietic potential produce the highest numbers of T cells when cultured on OP9-DLL4 as compared with downstream progenitors, including HE and multipotent hematopoietic progenitors. In addition, we found that T cells generated from HVMPs have the capacity to expand for 6-7 wk in vitro, in comparison with T cells generated from HE and hematopoietic progenitors, which could only be expanded for 4-5 wk. Demonstrating the critical need of NOTCH activation at the HVMP stage of hematopoietic development to establish robust T cell production from hPSCs may aid in establishing protocols for the efficient off-the-shelf production and expansion of T cells for treating hematologic malignancies.


Subject(s)
Cell Proliferation , Lymphopoiesis , Mesoderm/cytology , Pluripotent Stem Cells/cytology , Receptor, Notch1/genetics , T-Lymphocytes/cytology , Animals , Cell Line , Coculture Techniques , Fibroblasts , Flow Cytometry , Humans , Mice
6.
Cell Mol Life Sci ; 75(19): 3507-3520, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29992471

ABSTRACT

Mesenchymoangioblast (MB) is the earliest precursor for endothelial and mesenchymal cells originating from APLNR+PDGFRα+KDR+ mesoderm in human pluripotent stem cell cultures. MBs are identified based on their capacity to form FGF2-dependent compact spheroid colonies in a serum-free semisolid medium. MBs colonies are composed of PDGFRß+CD271+EMCN+DLK1+CD73- primitive mesenchymal cells which are generated through endothelial/angioblastic intermediates (cores) formed during first 3-4 days of clonogenic cultures. MB-derived primitive mesenchymal cells have potential to differentiate into mesenchymal stromal/stem cells (MSCs), pericytes, and smooth muscle cells. In this review, we summarize the specification and developmental potential of MBs, emphasize features that distinguish MBs from other mesenchymal progenitors described in the literature and discuss the value of these findings for identifying molecular pathways leading to MSC and vasculogenic cell specification, and developing cellular therapies using MB-derived progeny.


Subject(s)
Endothelial Cells/cytology , Mesenchymal Stem Cells/cytology , Mesoderm/cytology , Autoimmune Diseases/therapy , Cell Lineage , Embryonic Development , Endothelial Cells/metabolism , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mesoderm/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism
7.
Proc Natl Acad Sci U S A ; 113(51): E8257-E8266, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27930301

ABSTRACT

Artificial transcription factors (ATFs) are precision-tailored molecules designed to bind DNA and regulate transcription in a preprogrammed manner. Libraries of ATFs enable the high-throughput screening of gene networks that trigger cell fate decisions or phenotypic changes. We developed a genome-scale library of ATFs that display an engineered interaction domain (ID) to enable cooperative assembly and synergistic gene expression at targeted sites. We used this ATF library to screen for key regulators of the pluripotency network and discovered three combinations of ATFs capable of inducing pluripotency without exogenous expression of Oct4 (POU domain, class 5, TF 1). Cognate site identification, global transcriptional profiling, and identification of ATF binding sites reveal that the ATFs do not directly target Oct4; instead, they target distinct nodes that converge to stimulate the endogenous pluripotency network. This forward genetic approach enables cell type conversions without a priori knowledge of potential key regulators and reveals unanticipated gene network dynamics that drive cell fate choices.


Subject(s)
Cell Lineage , Cellular Reprogramming , Transcription Factors/metabolism , Animals , Binding Sites/genetics , Chaperonin Containing TCP-1/metabolism , Epigenesis, Genetic , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genomic Library , HEK293 Cells , Humans , Mice , Protein Domains , Protein Engineering , Sequence Analysis, RNA , Transcription Factors/genetics , Transcription, Genetic , Zinc Fingers/genetics
8.
Blood ; 122(25): 4035-46, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24124087

ABSTRACT

Significant advances in cellular reprogramming technologies and hematopoietic differentiation from human pluripotent stem cells (hPSCs) have already enabled the routine production of multiple lineages of blood cells in vitro and opened novel opportunities to study hematopoietic development, model genetic blood diseases, and manufacture immunologically matched cells for transfusion and cancer immunotherapy. However, the generation of hematopoietic cells with robust and sustained multilineage engraftment has not been achieved. Here, we highlight the recent advances in understanding the molecular and cellular pathways leading to blood development from hPSCs and discuss potential approaches that can be taken to facilitate the development of technologies for de novo production of hematopoietic stem cells.


Subject(s)
Cell Differentiation , Genetic Diseases, Inborn/metabolism , Hematologic Diseases/metabolism , Hematopoietic Stem Cells/metabolism , Neoplasms/metabolism , Pluripotent Stem Cells/metabolism , Animals , Disease Models, Animal , Genetic Diseases, Inborn/pathology , Genetic Diseases, Inborn/therapy , Hematologic Diseases/pathology , Hematologic Diseases/therapy , Humans , Neoplasms/pathology , Neoplasms/therapy , Stem Cell Transplantation/methods
9.
J Leukoc Biol ; 116(1): 118-131, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38417030

ABSTRACT

Neutrophils are rapidly recruited to sites of infection and are critical for pathogen clearance. Therapeutic use of primary neutrophils has been limited, as they have a short lifespan and are not amenable to genetic manipulation. Human induced pluripotent stem cells (iPSCs) can provide a robust source of neutrophils for infusion and are genetically tractable. However, current work has indicated that dampened intracellular signaling limits iPSC-derived neutrophil (iNeutrophil) cellular activation and antimicrobial response. Here, we show that protein tyrosine phosphatase 1B (PTP1B) inhibits intracellular signaling and dampens iNeutrophil effector function. Deletion of the PTP1B phosphatase increased PI3K and ERK signaling and was associated with increased F-actin polymerization, cell migration, and phagocytosis. In contrast, other effector functions like NETosis and reactive oxygen species production were reduced. PTP1B-deficient neutrophils were more responsive to Aspergillus fumigatus and displayed rapid recruitment and control of hyphal growth. Accordingly, depletion of PTP1B increased production of inflammatory factors including the neutrophil chemokine interleukin-8. Taken together, these findings suggest that PTP1B limits iNeutrophil motility and antimicrobial function.


Subject(s)
Cell Movement , Induced Pluripotent Stem Cells , Neutrophils , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Neutrophils/metabolism , Neutrophils/immunology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Aspergillus fumigatus , Phagocytosis , Phosphatidylinositol 3-Kinases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Extracellular Traps/metabolism , Extracellular Traps/immunology , Actins/metabolism
10.
Blood ; 117(14): e109-19, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21296996

ABSTRACT

Reprogramming blood cells to induced pluripotent stem cells (iPSCs) provides a novel tool for modeling blood diseases in vitro. However, the well-known limitations of current reprogramming technologies include low efficiency, slow kinetics, and transgene integration and residual expression. In the present study, we have demonstrated that iPSCs free of transgene and vector sequences could be generated from human BM and CB mononuclear cells using non-integrating episomal vectors. The reprogramming described here is up to 100 times more efficient, occurs 1-3 weeks faster compared with the reprogramming of fibroblasts, and does not require isolation of progenitors or multiple rounds of transfection. Blood-derived iPSC lines lacked rearrangements of IGH and TCR, indicating that their origin is non-B- or non-T-lymphoid cells. When cocultured on OP9, blood-derived iPSCs could be differentiated back to the blood cells, albeit with lower efficiency compared to fibroblast-derived iPSCs. We also generated transgene-free iPSCs from the BM of a patient with chronic myeloid leukemia (CML). CML iPSCs showed a unique complex chromosomal translocation identified in marrow sample while displaying typical embryonic stem cell phenotype and pluripotent differentiation potential. This approach provides an opportunity to explore banked normal and diseased CB and BM samples without the limitations associated with virus-based methods.


Subject(s)
Bone Marrow Cells/physiology , Bone Marrow Neoplasms/pathology , Cellular Reprogramming/physiology , Fetal Blood/cytology , Induced Pluripotent Stem Cells/physiology , Leukocytes, Mononuclear/physiology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Culture Techniques/methods , Cell Dedifferentiation/physiology , Cells, Cultured , Cellular Reprogramming/genetics , Coculture Techniques/methods , Efficiency , Fetal Blood/metabolism , Fetal Blood/physiology , Gene Expression Profiling , Gene Transfer Techniques , Humans , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Mice , Microarray Analysis , Transgenes/physiology
11.
Stem Cell Reports ; 18(2): 585-596, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36638788

ABSTRACT

Macrophages armed with chimeric antigen receptors (CARs) provide a potent new option for treating solid tumors. However, genetic engineering and scalable production of somatic macrophages remains significant challenges. Here, we used CRISPR-Cas9 gene editing methods to integrate an anti-GD2 CAR into the AAVS1 locus of human pluripotent stem cells (hPSCs). We then established a serum- and feeder-free differentiation protocol for generating CAR macrophages (CAR-Ms) through arterial endothelial-to-hematopoietic transition (EHT). CAR-M produced by this method displayed a potent cytotoxic activity against GD2-expressing neuroblastoma and melanoma in vitro and neuroblastoma in vivo. This study provides a new platform for the efficient generation of off-the-shelf CAR-Ms for antitumor immunotherapy.


Subject(s)
Melanoma , Neuroblastoma , Pluripotent Stem Cells , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/genetics , Receptors, Antigen, T-Cell/genetics , Immunotherapy/methods , Pluripotent Stem Cells/pathology , Melanoma/therapy , Neuroblastoma/therapy , Neuroblastoma/pathology , Macrophages/pathology
12.
iScience ; 26(5): 106621, 2023 May 19.
Article in English | MEDLINE | ID: mdl-37250328

ABSTRACT

Hemogenic endothelium (HE) is the main source of blood cells in the embryo. To improve blood manufacturing from human pluripotent stem cells (hPSCs), it is essential to define the molecular determinants that enhance HE specification and promote development of the desired blood lineage from HE. Here, using SOX18-inducible hPSCs, we revealed that SOX18 forced expression at the mesodermal stage, in contrast to its homolog SOX17, has minimal effects on arterial specification of HE, expression of HOXA genes and lymphoid differentiation. However, forced expression of SOX18 in HE during endothelial-to-hematopoietic transition (EHT) greatly increases NK versus T cell lineage commitment of hematopoietic progenitors (HPs) arising from HE predominantly expanding CD34+CD43+CD235a/CD41a-CD45- multipotent HPs and altering the expression of genes related to T cell and Toll-like receptor signaling. These studies improve our understanding of lymphoid cell specification during EHT and provide a new tool for enhancing NK cell production from hPSCs for immunotherapies.

13.
J Immunol ; 185(9): 5130-9, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20889540

ABSTRACT

Since the discovery that CXCR7 binds to CXCL12/SDF-1α, the role of CXCR7 in CXCL12-mediated biological processes has been under intensive scrutiny. However, there is no consensus in the literature on the expression of CXCR7 protein by peripheral blood cells. In this study we analyzed human and mouse leukocytes and erythrocytes for CXCR7 protein expression, using a competitive CXCL12 binding assay as well as by flow cytometry and immunohistochemistry using multiple CXCR7 Abs. CXCR7(-/-) mice were used as negative controls. Together, these methods indicate that CXCR7 protein is not expressed by human peripheral blood T cells, B cells, NK cells, or monocytes, or by mouse peripheral blood leukocytes. CXCR7 protein is, however, expressed on mouse primitive erythroid cells, which supply oxygen to the embryo during early stages of development. These studies therefore suggest that, whereas CXCR7 protein is expressed by primitive RBCs during murine embryonic development, in adult mammals CXCR7 protein is not expressed by normal peripheral blood cells.


Subject(s)
Erythrocytes/metabolism , Leukocytes/metabolism , Receptors, CXCR/biosynthesis , Adult , Animals , Cell Separation , Embryo, Mammalian , Flow Cytometry , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Immunohistochemistry , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
14.
Methods Mol Biol ; 2429: 103-124, 2022.
Article in English | MEDLINE | ID: mdl-35507158

ABSTRACT

Endothelial-to-hematopoietic transition (EHT) is a unique morphogenic event in which flat, adherent hemogenic endothelial (HE) cells acquire round, non-adherent blood cell morphology. Investigating the mechanisms of EHT is critical for understanding the development of hematopoietic stem cells (HSCs) and the entirety of the adult immune system, and advancing technologies for manufacturing blood cells from human pluripotent stem cells (hPSCs). Here we describe a protocol to (a) generate and isolate subsets of HE from hPSCs, (b) assess EHT and hematopoietic potential of HE subsets in bulk cultures and at the single-cell level, and (c) evaluate the role of NOTCH signaling during HE specification and EHT. The generation of HE from hPSCs and EHT bulk cultures are performed in xenogen- and feeder-free system, providing the unique advantage of being able to investigate the role of individual signaling factors during EHT and the definitive lympho-myeloid cell specification from hPSCs.


Subject(s)
Hemangioblasts , Pluripotent Stem Cells , Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cells , Humans
15.
Front Genome Ed ; 4: 1031275, 2022.
Article in English | MEDLINE | ID: mdl-36714391

ABSTRACT

Introduction: Genome editing by CRISPR-Cas9 approaches offers promise for introducing or correcting disease-associated mutations for research and clinical applications. Nonhuman primates are physiologically closer to humans than other laboratory animal models, providing ideal candidates for introducing human disease-associated mutations to develop models of human disease. The incidence of large chromosomal anomalies in CRISPR-Cas9-edited human embryos and cells warrants comprehensive genotypic investigation of editing outcomes in primate embryos. Our objective was to evaluate on- and off-target editing outcomes in CCR5 CRISPR-Cas9-targeted Mauritian cynomolgus macaque embryos. Methods: DNA isolated from individual blastomeres of two embryos, along with paternal and maternal DNA, was subjected to whole genome sequencing (WGS) analysis. Results: Large deletions were identified in macaque blastomeres at the on-target site that were not previously detected using PCR-based methods. De novo mutations were also identified at predicted CRISPR-Cas9 off-target sites. Discussion: This is the first report of WGS analysis of CRISPR-Cas9-targeted nonhuman primate embryonic cells, in which a high editing efficiency was coupled with the incidence of editing errors in cells from two embryos. These data demonstrate that comprehensive sequencing-based methods are warranted for evaluating editing outcomes in primate embryos, as well as any resultant offspring to ensure that the observed phenotype is due to the targeted edit and not due to unidentified off-target mutations.

16.
Stem Cell Reports ; 17(4): 953-963, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35364011

ABSTRACT

Adoptive therapies with genetically modified somatic T cells rendered HIV resistance have shown promise for AIDS therapy. A renewable source of HIV-resistant human T cells from induced pluripotent stem cells (iPSCs) would further facilitate and broaden the applicability of these therapies. Here, we report successful targeting of the CCR5 locus in iPSCs generated from T cells (T-iPSCs) or fibroblasts (fib-iPSCs) from Mauritian cynomolgus macaques (MCM), using CRISPR-Cas9 technology. We found that CCR5 editing does not affect hematopoietic and T cell differentiation potentials of fib-iPSCs. However, T-iPSCs with edited CCR5 lost their capacity to differentiate into CD4+CD8+ T cells while maintaining myeloid differentiation potential. T cells and macrophages produced from CCR5-edited MCM iPSCs did not support replication of the CCR5-tropic simian immunodeficiency viruses SIVmac239 (T cell tropic) and SIVmac316 (macrophage-tropic). Overall, these studies provide a platform for further exploration of AIDS therapies based on gene-edited iPSCs in a nonhuman primate model.


Subject(s)
Acquired Immunodeficiency Syndrome , Induced Pluripotent Stem Cells , Animals , CD8-Positive T-Lymphocytes , Macaca fascicularis , Macrophages , Receptors, CCR5/genetics
17.
Blood Adv ; 6(18): 5267-5278, 2022 09 27.
Article in English | MEDLINE | ID: mdl-35404997

ABSTRACT

Administration of ex vivo expanded somatic myeloid progenitors has been explored as a way to facilitate a more rapid myeloid recovery and improve overall survival after myeloablation. Recent advances in induced pluripotent stem cell (iPSC) technologies have created alternative platforms for supplying off-the-shelf immunologically compatible myeloid progenitors, including cellular products derived from major histocompatibility complex (MHC) homozygous superdonors, potentially increasing the availability of MHC-matching cells and maximizing the utility of stem cell banking. However, the teratogenic and tumorigenic potential of iPSC-derived progenitor cells and whether they will induce alloreactive antibodies upon transfer remain unclear. We evaluated the safety and efficacy of using CD34+CD45+ hematopoietic progenitors derived from MHC homozygous iPSCs (iHPs) to treat cytopenia after myeloablative hematopoietic stem cell (HSC) transplantation in a Mauritian cynomolgus macaque (MCM) nonhuman primate (NHP) model. We demonstrated that infusion of iHPs was well tolerated and safe, observing no teratomas or tumors in the MCMs up to 1 year after HSC transplantation and iHP infusion. Importantly, the iHPs also did not induce significant levels of alloantibodies in MHC-matched or -mismatched immunocompetent MCMs, even after increasing MHC expression on iHPs with interferon-γ. These results support the feasibility of iHP use in the setting of myeloablation and suggest that iHP products pose a low risk of inducing alloreactive antibodies.


Subject(s)
Hematopoietic Stem Cell Transplantation , Induced Pluripotent Stem Cells , Animals , Antigens, CD34 , Interferon-gamma , Isoantibodies , Macaca fascicularis , Major Histocompatibility Complex
18.
Sci Rep ; 12(1): 12345, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35853970

ABSTRACT

Allogeneic hematopoietic stem cell transplants (allo-HSCTs) dramatically reduce HIV reservoirs in antiretroviral therapy (ART) suppressed individuals. However, the mechanism(s) responsible for these post-transplant viral reservoir declines are not fully understood. Therefore, we modeled allo-HSCT in ART-suppressed simian-human immunodeficiency virus (SHIV)-infected Mauritian cynomolgus macaques (MCMs) to illuminate factors contributing to transplant-induced viral reservoir decay. Thus, we infected four MCMs with CCR5-tropic SHIV162P3 and started them on ART 6-16 weeks post-infection (p.i.), maintaining continuous ART during myeloablative conditioning. To prevent graft-versus-host disease (GvHD), we transplanted allogeneic MHC-matched α/ß T cell-depleted bone marrow cells and prophylactically treated the MCMs with cyclophosphamide and tacrolimus. The transplants produced ~ 85% whole blood donor chimerism without causing high-grade GvHD. Consequently, three MCMs had undetectable SHIV DNA in their blood post-transplant. However, SHIV-harboring cells persisted in various tissues, with detectable viral DNA in lymph nodes and tissues between 38 and 62 days post-transplant. Further, removing one MCM from ART at 63 days post-transplant resulted in SHIV rapidly rebounding within 7 days of treatment withdrawal. In conclusion, transplanting SHIV-infected MCMs with allogeneic MHC-matched α/ß T cell-depleted bone marrow cells prevented high-grade GvHD and decreased SHIV-harboring cells in the blood post-transplant but did not eliminate viral reservoirs in tissues.


Subject(s)
Graft vs Host Disease , HIV Infections , Hematopoietic Stem Cell Transplantation , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Bone Marrow Transplantation/adverse effects , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , HIV , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Macaca fascicularis , Receptors, Antigen, T-Cell , Simian Immunodeficiency Virus/genetics
19.
Cell Rep ; 34(7): 108758, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33596423

ABSTRACT

SOX17 has been implicated in arterial specification and the maintenance of hematopoietic stem cells (HSCs) in the murine embryo. However, knowledge about molecular pathways and stage-specific effects of SOX17 in humans remains limited. Here, using SOX17-knockout and SOX17-inducible human pluripotent stem cells (hPSCs), paired with molecular profiling studies, we reveal that SOX17 is a master regulator of HOXA and arterial programs in hemogenic endothelium (HE) and is required for the specification of HE with robust lympho-myeloid potential and DLL4+CXCR4+ phenotype resembling arterial HE at the sites of HSC emergence. Along with the activation of NOTCH signaling, SOX17 directly activates CDX2 expression, leading to the upregulation of the HOXA cluster genes. Since deficiencies in HOXA and NOTCH signaling contribute to the impaired in vivo engraftment of hPSC-derived hematopoietic cells, the identification of SOX17 as a key regulator linking arterial and HOXA programs in HE may help to program HSC fate from hPSCs.


Subject(s)
Hematopoiesis/genetics , Homeodomain Proteins/metabolism , SOXF Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism
20.
Exp Hematol ; 93: 44-51, 2021 01.
Article in English | MEDLINE | ID: mdl-33176119

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (alloHSCT) is a potentially curative treatment for hematologic cancers and chronic infections such as human immunodeficiency virus (HIV). Its success in these settings is attributed to the ability of engrafting immune cells to eliminate cancer cells or deplete the HIV reservoir (graft-versus-host effect [GvHE]). However, alloHSCT is commonly associated with graft-versus-host diseases (GvHDs) causing significant morbidity and mortality, thereby requiring development of novel allogeneic HSCT protocols and therapies promoting GvHE without GvHD using physiologically relevant preclinical models. Here we evaluated the outcomes of major histocompatibility complex-matched T-cell receptor α/ß-depleted alloHSCT in Mauritian cynomolgus macaques (MCMs). Following T-cell receptor α/ß depletion, bone marrow cells were transplanted into major histocompatibility complex-identical MCMs conditioned with total body irradiation. GvHD prophylaxis included sirolimus alone in two animals or tacrolimus with cyclophosphamide in another two animals. Posttransplant chimerism was determined by sequencing diagnostic single-nucleotide polymorphisms to quantify the amounts of donor and recipient cells present in blood. Animals treated posttransplant with sirolimus developed nearly complete chimerism with acute GvHD. In the cyclophosphamide and tacrolimus treatment group, animals developed mixed chimerism without GvHD, with long-term engraftment observed in one animal. None of the animals developed cytomegalovirus infection. These studies indicate the feasibility of alloHSCT engraftment without GvHD in an MHC-identical MCM model following complete myeloablative conditioning and anti-GvHD prophylaxis with posttransplant cyclophosphamide and tacrolimus. Further exploration of this model will provide a platform for elucidating the mechanisms of GvHD and GvHE and for testing novel alloHSCT modalities for HIV infection.


Subject(s)
Bone Marrow Cells/cytology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/methods , Receptors, Antigen, T-Cell/isolation & purification , Animals , Female , Hematopoietic Stem Cells/cytology , Immunosuppressive Agents/therapeutic use , Macaca fascicularis , Male , Sirolimus/therapeutic use , Transplantation Conditioning/methods , Transplantation, Homologous/methods , Whole-Body Irradiation/methods
SELECTION OF CITATIONS
SEARCH DETAIL