Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4325, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773071

ABSTRACT

Hematopoietic stem cell (HSC) mutations can result in clonal hematopoiesis (CH) with heterogeneous clinical outcomes. Here, we investigate how the cell state preceding Tet2 mutation impacts the pre-malignant phenotype. Using an inducible system for clonal analysis of myeloid progenitors, we find that the epigenetic features of clones at similar differentiation status are highly heterogeneous and functionally respond differently to Tet2 mutation. Cell differentiation stage also influences Tet2 mutation response indicating that the cell of origin's epigenome modulates clone-specific behaviors in CH. Molecular features associated with higher risk outcomes include Sox4 that sensitizes cells to Tet2 inactivation, inducing dedifferentiation, altered metabolism and increasing the in vivo clonal output of mutant cells, as confirmed in primary GMP and HSC models. Our findings validate the hypothesis that epigenetic features can predispose specific clones for dominance, explaining why identical genetic mutations can result in different phenotypes.


Subject(s)
DNA-Binding Proteins , Dioxygenases , Epigenesis, Genetic , Hematopoietic Stem Cells , Mutation , Proto-Oncogene Proteins , Dioxygenases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Animals , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Humans , Hematopoiesis/genetics , Mice , Cell Differentiation/genetics
2.
Blood Adv ; 7(21): 6608-6623, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37450380

ABSTRACT

Myelodysplastic syndromes (MDSs) are a heterogenous group of diseases affecting the hematopoietic stem cell that are curable only by stem cell transplantation. Both hematopoietic cell intrinsic changes and extrinsic signals from the bone marrow (BM) niche seem to ultimately lead to MDS. Animal models of MDS indicate that alterations in specific mesenchymal progenitor subsets in the BM microenvironment can induce or select for abnormal hematopoietic cells. Here, we identify a subset of human BM mesenchymal cells marked by the expression of CD271, CD146, and CD106. This subset of human mesenchymal cells is comparable with mouse mesenchymal cells that, when perturbed, result in an MDS-like syndrome. Its transcriptional analysis identified Osteopontin (SPP1) as the most overexpressed gene. Selective depletion of Spp1 in the microenvironment of the mouse MDS model, Vav-driven Nup98-HoxD13, resulted in an accelerated progression as demonstrated by increased chimerism, higher mutant myeloid cell burden, and a more pronounced anemia when compared with that in wild-type microenvironment controls. These data indicate that molecular perturbations can occur in specific BM mesenchymal subsets of patients with MDS. However, the niche adaptations to dysplastic clones include Spp1 overexpression that can constrain disease fitness and potentially progression. Therefore, niche changes with malignant disease can also serve to protect the host.


Subject(s)
Bone Marrow , Myelodysplastic Syndromes , Humans , Mice , Animals , Bone Marrow/pathology , Myelodysplastic Syndromes/genetics , Hematopoietic Stem Cells/metabolism , Bone Marrow Cells/metabolism , Disease Models, Animal , Disease Progression
3.
Blood ; 142(7): 658-674, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37267513

ABSTRACT

Myeloid cell heterogeneity is known, but whether it is cell-intrinsic or environmentally-directed remains unclear. Here, an inducible/reversible system pausing myeloid differentiation allowed the definition of clone-specific functions that clustered monocytes into subsets with distinctive molecular features. These subsets were orthogonal to the classical/nonclassical categorization and had inherent, restricted characteristics that did not shift under homeostasis, after irradiation, or with infectious stress. Rather, their functional fate was constrained by chromatin accessibility established at or before the granulocyte-monocyte or monocyte-dendritic progenitor level. Subsets of primary monocytes had differential ability to control distinct infectious agents in vivo. Therefore, monocytes are a heterogeneous population of functionally restricted subtypes defined by the epigenome of their progenitors that are differentially selected by physiologic challenges with limited plasticity to transition from one subset to another.


Subject(s)
Granulocytes , Monocytes , Myeloid Progenitor Cells , Epigenome , Epigenesis, Genetic , Cell Differentiation/genetics
4.
Elife ; 112022 11 16.
Article in English | MEDLINE | ID: mdl-36382886

ABSTRACT

A major goal of biological imaging is localization of biomolecules inside a cell. Fluorescence microscopy can localize biomolecules inside whole cells and tissues, but its ability to count biomolecules and accuracy of the spatial coordinates is limited by the wavelength of visible light. Cryo-electron microscopy (cryo-EM) provides highly accurate position and orientation information of biomolecules but is often confined to small fields of view inside a cell, limiting biological context. In this study, we use a new data-acquisition scheme called Defocus-Corrected Large-Area cryo-EM (DeCo-LACE) to collect high-resolution images of entire sections (100- to 250-nm-thick lamellae) of neutrophil-like mouse cells, representing 1-2% of the total cellular volume. We use 2D template matching (2DTM) to determine localization and orientation of the large ribosomal subunit in these sections. These data provide maps of ribosomes across entire sections of mammalian cells. This high-throughput cryo-EM data collection approach together with 2DTM will advance visual proteomics and provide biological insight that cannot be obtained by other methods.


Subject(s)
Mammals , Ribosomes , Animals , Mice , Cryoelectron Microscopy/methods , Microscopy, Fluorescence/methods
5.
J Exp Med ; 219(11)2022 11 07.
Article in English | MEDLINE | ID: mdl-36129517

ABSTRACT

A sleepless night may feel awful in its aftermath, but sleep's revitalizing powers are substantial, perpetuating the idea that convalescent sleep is a consequence-free physiological reset. Although recent studies have shown that catch-up sleep insufficiently neutralizes the negative effects of sleep debt, the mechanisms that control prolonged effects of sleep disruption are not understood. Here, we show that sleep interruption restructures the epigenome of hematopoietic stem and progenitor cells (HSPCs) and increases their proliferation, thus reducing hematopoietic clonal diversity through accelerated genetic drift. Sleep fragmentation exerts a lasting influence on the HSPC epigenome, skewing commitment toward a myeloid fate and priming cells for exaggerated inflammatory bursts. Combining hematopoietic clonal tracking with mathematical modeling, we infer that sleep preserves clonal diversity by limiting neutral drift. In humans, sleep restriction alters the HSPC epigenome and activates hematopoiesis. These findings show that sleep slows decay of the hematopoietic system by calibrating the hematopoietic epigenome, constraining inflammatory output, and maintaining clonal diversity.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells , Cells, Cultured , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , Humans , Sleep/genetics
6.
Nat Med ; 27(8): 1458-1470, 2021 08.
Article in English | MEDLINE | ID: mdl-34140705

ABSTRACT

Gene therapy (GT) has rapidly attracted renewed interest as a treatment for otherwise incurable diseases, with several GT products already on the market and many more entering clinical testing for selected indications. Clonal tracking techniques based on vector integration enable monitoring of the fate of engineered cells in the blood of patients receiving GT and allow assessment of the safety and efficacy of these procedures. However, owing to the limited number of cells that can be tested and the impracticality of studying cells residing in peripheral organs without performing invasive biopsies, this approach provides only a partial snapshot of the clonal repertoire and dynamics of genetically modified cells and reduces the predictive power as a safety readout. In this study, we developed liquid biopsy integration site sequencing, or LiBIS-seq, a polymerase chain reaction technique optimized to quantitatively retrieve vector integration sites from cell-free DNA released into the bloodstream by dying cells residing in several tissues. This approach enabled longitudinal monitoring of in vivo liver-directed GT and clonal tracking in patients receiving hematopoietic stem cell GT, improving our understanding of the clonal composition and turnover of genetically modified cells in solid tissues and, in contrast to conventional analyses based only on circulating blood cells, enabling earlier detection of vector-marked clones that are aberrantly expanding in peripheral tissues.


Subject(s)
Cell-Free Nucleic Acids/genetics , Genetic Vectors/genetics , Cell-Free Nucleic Acids/adverse effects , Genetic Therapy , Humans , Leukemia/genetics , Leukemia/therapy , Leukodystrophy, Metachromatic/genetics , Leukodystrophy, Metachromatic/therapy , Lymphoma/genetics , Lymphoma/therapy
7.
EMBO Mol Med ; 13(3): e13545, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33475257

ABSTRACT

Precise correction of the CD40LG gene in T cells and hematopoietic stem/progenitor cells (HSPC) holds promise for treating X-linked hyper-IgM Syndrome (HIGM1), but its actual therapeutic potential remains elusive. Here, we developed a one-size-fits-all editing strategy for effective T-cell correction, selection, and depletion and investigated the therapeutic potential of T-cell and HSPC therapies in the HIGM1 mouse model. Edited patients' derived CD4 T cells restored physiologically regulated CD40L expression and contact-dependent B-cell helper function. Adoptive transfer of wild-type T cells into conditioned HIGM1 mice rescued antigen-specific IgG responses and protected mice from a disease-relevant pathogen. We then obtained ~ 25% CD40LG editing in long-term repopulating human HSPC. Transplanting such proportion of wild-type HSPC in HIGM1 mice rescued immune functions similarly to T-cell therapy. Overall, our findings suggest that autologous edited T cells can provide immediate and substantial benefits to HIGM1 patients and position T-cell ahead of HSPC gene therapy because of easier translation, lower safety concerns and potentially comparable clinical benefits.


Subject(s)
Hyper-IgM Immunodeficiency Syndrome, Type 1 , Hyper-IgM Immunodeficiency Syndrome , Animals , Gene Editing , Hematopoietic Stem Cells , Humans , Mice , T-Lymphocytes
8.
Cell Stem Cell ; 24(4): 551-565.e8, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30905619

ABSTRACT

Precise gene editing in hematopoietic stem and progenitor cells (HSPCs) holds promise for treating genetic diseases. However, responses triggered by programmable nucleases in HSPCs are poorly characterized and may negatively impact HSPC engraftment and long-term repopulation capacity. Here, we induced either one or several DNA double-stranded breaks (DSBs) with optimized zinc-finger and CRISPR/Cas9 nucleases and monitored DNA damage response (DDR) foci induction, cell-cycle progression, and transcriptional responses in HSPC subpopulations, with up to single-cell resolution. p53-mediated DDR pathway activation was the predominant response to even single-nuclease-induced DSBs across all HSPC subtypes analyzed. Excess DSB load and/or adeno-associated virus (AAV)-mediated delivery of DNA repair templates induced cumulative p53 pathway activation, constraining proliferation, yield, and engraftment of edited HSPCs. However, functional impairment was reversible when DDR burden was low and could be overcome by transient p53 inhibition. These findings provide molecular and functional evidence for feasible and seamless gene editing in HSPCs.


Subject(s)
DNA Damage , Gene Editing , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Humans , K562 Cells , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID
9.
Cell Stem Cell ; 23(6): 820-832.e9, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30416070

ABSTRACT

Innate immune factors may restrict hematopoietic stem cell (HSC) genetic engineering and contribute to broad individual variability in gene therapy outcomes. Here, we show that HSCs harbor an early, constitutively active innate immune block to lentiviral transduction that can be efficiently overcome by cyclosporine H (CsH). CsH potently enhances gene transfer and editing in human long-term repopulating HSCs by inhibiting interferon-induced transmembrane protein 3 (IFITM3), which potently restricts VSV glycoprotein-mediated vector entry. Importantly, individual variability in endogenous IFITM3 levels correlated with permissiveness of HSCs to lentiviral transduction, suggesting that CsH treatment will be useful for improving ex vivo gene therapy and standardizing HSC transduction across patients. Overall, our work unravels the involvement of innate pathogen recognition molecules in immune blocks to gene correction in primary human HSCs and highlights how these roadblocks can be overcome to develop innovative cell and gene therapies.


Subject(s)
Cyclosporine/pharmacology , Gene Editing , Hematopoietic Stem Cells/drug effects , Immunity, Innate/drug effects , Lentivirus/drug effects , Lentivirus/genetics , Transduction, Genetic , Animals , Cell Line , Female , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Humans , Lentivirus/immunology , Mice , Mice, Inbred NOD , Mice, Knockout
10.
Sci Transl Med ; 9(411)2017 Oct 11.
Article in English | MEDLINE | ID: mdl-29021165

ABSTRACT

Targeted genome editing in hematopoietic stem/progenitor cells (HSPCs) is an attractive strategy for treating immunohematological diseases. However, the limited efficiency of homology-directed editing in primitive HSPCs constrains the yield of corrected cells and might affect the feasibility and safety of clinical translation. These concerns need to be addressed in stringent preclinical models and overcome by developing more efficient editing methods. We generated a humanized X-linked severe combined immunodeficiency (SCID-X1) mouse model and evaluated the efficacy and safety of hematopoietic reconstitution from limited input of functional HSPCs, establishing thresholds for full correction upon different types of conditioning. Unexpectedly, conditioning before HSPC infusion was required to protect the mice from lymphoma developing when transplanting small numbers of progenitors. We then designed a one-size-fits-all IL2RG (interleukin-2 receptor common γ-chain) gene correction strategy and, using the same reagents suitable for correction of human HSPC, validated the edited human gene in the disease model in vivo, providing evidence of targeted gene editing in mouse HSPCs and demonstrating the functionality of the IL2RG-edited lymphoid progeny. Finally, we optimized editing reagents and protocol for human HSPCs and attained the threshold of IL2RG editing in long-term repopulating cells predicted to safely rescue the disease, using clinically relevant HSPC sources and highly specific zinc finger nucleases or CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 (CRISPR-associated protein 9). Overall, our work establishes the rationale and guiding principles for clinical translation of SCID-X1 gene editing and provides a framework for developing gene correction for other diseases.


Subject(s)
Hematopoietic Stem Cells/metabolism , Animals , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing/methods , Gene Targeting/methods , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/metabolism , Mice , Mice, SCID
11.
EMBO Mol Med ; 9(11): 1574-1588, 2017 11.
Article in English | MEDLINE | ID: mdl-28899930

ABSTRACT

Gene targeting constitutes a new step in the development of gene therapy for inherited diseases. Although previous studies have shown the feasibility of editing fibroblasts from Fanconi anemia (FA) patients, here we aimed at conducting therapeutic gene editing in clinically relevant cells, such as hematopoietic stem cells (HSCs). In our first experiments, we showed that zinc finger nuclease (ZFN)-mediated insertion of a non-therapeutic EGFP-reporter donor in the AAVS1 "safe harbor" locus of FA-A lymphoblastic cell lines (LCLs), indicating that FANCA is not essential for the editing of human cells. When the same approach was conducted with therapeutic FANCA donors, an efficient phenotypic correction of FA-A LCLs was obtained. Using primary cord blood CD34+ cells from healthy donors, gene targeting was confirmed not only in in vitro cultured cells, but also in hematopoietic precursors responsible for the repopulation of primary and secondary immunodeficient mice. Moreover, when similar experiments were conducted with mobilized peripheral blood CD34+ cells from FA-A patients, we could demonstrate for the first time that gene targeting in primary hematopoietic precursors from FA patients is feasible and compatible with the phenotypic correction of these clinically relevant cells.


Subject(s)
Antigens, CD34/metabolism , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia/pathology , Gene Editing/methods , Hematopoietic Stem Cells/metabolism , Animals , Base Sequence , Cells, Cultured , Dependovirus/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein/metabolism , Fetal Blood/cytology , Genetic Vectors/genetics , Genetic Vectors/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Reactive Oxygen Species/metabolism , Zinc Finger Nucleases/genetics , Zinc Finger Nucleases/metabolism
12.
Blood ; 130(5): 606-618, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28637663

ABSTRACT

Transfer of T-cell receptors (TCRs) specific for tumor-associated antigens is a promising approach for cancer immunotherapy. We developed the TCR gene editing technology that is based on the knockout of the endogenous TCR α and ß genes, followed by the introduction of tumor-specific TCR genes, and that proved safer and more effective than conventional TCR gene transfer. Although successful, complete editing requires extensive cell manipulation and 4 transduction procedures. Here we propose a novel and clinically feasible TCR "single editing" (SE) approach, based on the disruption of the endogenous TCR α chain only, followed by the transfer of genes encoding for a tumor-specific TCR. We validated SE with the clinical grade HLA-A2 restricted NY-ESO-1157-165-specific TCR. SE allowed the rapid production of high numbers of tumor-specific T cells, with optimal TCR expression and preferential stem memory and central memory phenotype. Similarly to unedited T cells redirected by TCR gene transfer (TCR transferred [TR]), SE T cells efficiently killed NY-ESO-1pos targets; however, although TR cells proved highly alloreactive, SE cells showed a favorable safety profile. Accordingly, when infused in NSG mice previously engrafted with myeloma, SE cells mediated tumor rejection without inducing xenogeneic graft-versus-host disease, thus resulting in significantly higher survival than that observed in mice treated with TR cells. Overall, single TCR gene editing represents a clinically feasible approach that is able to increase the safety and efficacy of cancer adoptive immunotherapy.


Subject(s)
Adoptive Transfer , Gene Editing/methods , Immunologic Memory , Multiple Myeloma , Neoplasm Proteins , Peptide Fragments , Receptors, Antigen, T-Cell , T-Lymphocytes , Animals , Cell Line, Tumor , Female , Gene Transfer Techniques , Graft vs Host Disease , Mice , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Peptide Fragments/genetics , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Xenograft Model Antitumor Assays
13.
Nature ; 510(7504): 235-240, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24870228

ABSTRACT

Targeted genome editing by artificial nucleases has brought the goal of site-specific transgene integration and gene correction within the reach of gene therapy. However, its application to long-term repopulating haematopoietic stem cells (HSCs) has remained elusive. Here we show that poor permissiveness to gene transfer and limited proficiency of the homology-directed DNA repair pathway constrain gene targeting in human HSCs. By tailoring delivery platforms and culture conditions we overcame these barriers and provide stringent evidence of targeted integration in human HSCs by long-term multilineage repopulation of transplanted mice. We demonstrate the therapeutic potential of our strategy by targeting a corrective complementary DNA into the IL2RG gene of HSCs from healthy donors and a subject with X-linked severe combined immunodeficiency (SCID-X1). Gene-edited HSCs sustained normal haematopoiesis and gave rise to functional lymphoid cells that possess a selective growth advantage over those carrying disruptive IL2RG mutations. These results open up new avenues for treating SCID-X1 and other diseases.


Subject(s)
Gene Targeting/methods , Genome, Human/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Targeted Gene Repair/methods , X-Linked Combined Immunodeficiency Diseases/genetics , Animals , Antigens, CD34/metabolism , DNA, Complementary/genetics , Endonucleases/metabolism , Fetal Blood/cytology , Fetal Blood/metabolism , Fetal Blood/transplantation , Hematopoiesis/genetics , Hematopoietic Stem Cell Transplantation , Humans , Interleukin Receptor Common gamma Subunit/genetics , Male , Mice , Mutation/genetics , X-Linked Combined Immunodeficiency Diseases/therapy
SELECTION OF CITATIONS
SEARCH DETAIL