Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Ther Methods Clin Dev ; 32(1): 101183, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38282895

ABSTRACT

Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is a monogenic disorder caused by mutations in the FOXP3 gene, required for generation of regulatory T (Treg) cells. Loss of Treg cells leads to immune dysregulation characterized by multi-organ autoimmunity and early mortality. Hematopoietic stem cell (HSC) transplantation can be curative, but success is limited by autoimmune complications, donor availability and/or graft-vs.-host disease. Correction of FOXP3 in autologous HSC utilizing a homology-directed repair (HDR)-based platform may provide a safer alternative therapy. Here, we demonstrate efficient editing of FOXP3 utilizing co-delivery of Cas9 ribonucleoprotein complexes and adeno-associated viral vectors to achieve HDR rates of >40% in vitro using mobilized CD34+ cells from multiple donors. Using this approach to deliver either a GFP or a FOXP3 cDNA donor cassette, we demonstrate sustained bone marrow engraftment of approximately 10% of HDR-edited cells in immune-deficient recipient mice at 16 weeks post-transplant. Further, we show targeted integration of FOXP3 cDNA in CD34+ cells from an IPEX patient and expression of the introduced FOXP3 transcript in gene-edited primary T cells from both healthy individuals and IPEX patients. Our combined findings suggest that refinement of this approach is likely to provide future clinical benefit in IPEX.

2.
Nat Commun ; 13(1): 6110, 2022 10 16.
Article in English | MEDLINE | ID: mdl-36245034

ABSTRACT

Due to their unique longevity and capacity to secrete high levels of protein, plasma B cells have the potential to be used as a cell therapy for protein replacement. Here, we show that ex vivo engineered human plasma cells exhibit single-cell RNA profiles, scanning electron micrograph ultrastructural features, and in vivo homing capacity of long-lived plasma cells. After transferring human plasma cells to immunodeficient mice in the presence of the human cytokines BAFF and IL-6, we observe increases in retention of plasma cells in the bone marrow, with engraftment exceeding a year. The most profound in vivo effects of human IL-6 are observed within 20 days of transfer and could be explained by decreased apoptosis in newly differentiated plasma cells. Collectively, these results show that ex vivo engineered and differentiated human plasma cells have the potential for long-lived in vivo protein secretion, which can be modeled in small animals.


Subject(s)
Hematopoietic Stem Cell Transplantation , Plasma Cells , Animals , Blood Proteins , Cytokines/metabolism , Humans , Interleukin-6 , Mice , Mice, SCID , Plasma Cells/metabolism , RNA
3.
Mol Ther Methods Clin Dev ; 20: 635-651, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33718514

ABSTRACT

X-linked agammaglobulinemia (XLA) is an immune disorder caused by mutations in Bruton's tyrosine kinase (BTK). BTK is expressed in B and myeloid cells, and its deficiency results in a lack of mature B cells and protective antibodies. We previously reported a lentivirus (LV) BTK replacement therapy that restored B cell development and function in Btk and Tec double knockout mice (a phenocopy of human XLA). In this study, with the goal of optimizing both the level and lineage specificity of BTK expression, we generated LV incorporating the proximal human BTK promoter. Hematopoietic stem cells from Btk -/- Tec -/- mice transduced with this vector rescued lineage-specific expression and restored B cell function in Btk -/- Tec -/- recipients. Next, we tested addition of candidate enhancers and/or ubiquitous chromatin opening elements (UCOEs), as well as codon optimization to improve BTK expression. An Eµ enhancer improved B cell rescue, but increased immunoglobulin G (IgG) autoantibodies. Addition of the UCOE avoided autoantibody generation while improving B cell development and function and reducing vector silencing. An optimized vector containing a truncated UCOE upstream of the BTK promoter and codon-optimized BTK cDNA resulted in stable, lineage-regulated BTK expression that mirrored endogenous BTK, making it a strong candidate for XLA therapy.

4.
Mol Ther ; 26(2): 456-467, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29273498

ABSTRACT

The ability to engineer primary human B cells to differentiate into long-lived plasma cells and secrete a de novo protein may allow the creation of novel plasma cell therapies for protein deficiency diseases and other clinical applications. We initially developed methods for efficient genome editing of primary B cells isolated from peripheral blood. By delivering CRISPR/CRISPR-associated protein 9 (Cas9) ribonucleoprotein (RNP) complexes under conditions of rapid B cell expansion, we achieved site-specific gene disruption at multiple loci in primary human B cells (with editing rates of up to 94%). We used this method to alter ex vivo plasma cell differentiation by disrupting developmental regulatory genes. Next, we co-delivered RNPs with either a single-stranded DNA oligonucleotide or adeno-associated viruses containing homologous repair templates. Using either delivery method, we achieved targeted sequence integration at high efficiency (up to 40%) via homology-directed repair. This method enabled us to engineer plasma cells to secrete factor IX (FIX) or B cell activating factor (BAFF) at high levels. Finally, we show that introduction of BAFF into plasma cells promotes their engraftment into immunodeficient mice. Our results highlight the utility of genome editing in studying human B cell biology and demonstrate a novel strategy for modifying human plasma cells to secrete therapeutic proteins.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Gene Editing , Genetic Engineering , Plasma Cells/immunology , Plasma Cells/metabolism , Recombinational DNA Repair , Animals , Biomarkers , CRISPR-Associated Protein 9 , Cytokines/metabolism , Dependovirus/genetics , Genetic Loci , Genetic Vectors/genetics , Humans , Immunotherapy , Mice , Phenotype , Polymorphism, Single Nucleotide , Positive Regulatory Domain I-Binding Factor 1/genetics , Receptors, CCR5/genetics , Transduction, Genetic
5.
Mol Ther ; 24(9): 1570-80, 2016 09 29.
Article in English | MEDLINE | ID: mdl-27203437

ABSTRACT

Many future therapeutic applications of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 and related RNA-guided nucleases are likely to require their use to promote gene targeting, thus necessitating development of methods that provide for delivery of three components-Cas9, guide RNAs and recombination templates-to primary cells rendered proficient for homology-directed repair. Here, we demonstrate an electroporation/transduction codelivery method that utilizes mRNA to express both Cas9 and mutant adenoviral E4orf6 and E1b55k helper proteins in association with adeno-associated virus (AAV) vectors expressing guide RNAs and recombination templates. By transiently enhancing target cell permissiveness to AAV transduction and gene editing efficiency, this novel approach promotes efficient gene disruption and/or gene targeting at multiple loci in primary human T-cells, illustrating its broad potential for application in translational gene editing.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Mutant Proteins , T-Lymphocytes/metabolism , Viral Proteins/metabolism , Dependovirus/genetics , Gene Expression , Gene Knock-In Techniques , Gene Knockout Techniques , Gene Order , Gene Targeting , Gene Transfer Techniques , Genetic Vectors/genetics , Homologous Recombination , Humans , RNA, Guide, Kinetoplastida/genetics , Transduction, Genetic , Viral Proteins/genetics
6.
Sci Transl Med ; 7(307): 307ra156, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26424571

ABSTRACT

Genetic mutations or engineered nucleases that disrupt the HIV co-receptor CCR5 block HIV infection of CD4(+) T cells. These findings have motivated the engineering of CCR5-specific nucleases for application as HIV therapies. The efficacy of this approach relies on efficient biallelic disruption of CCR5, and the ability to efficiently target sequences that confer HIV resistance to the CCR5 locus has the potential to further improve clinical outcomes. We used RNA-based nuclease expression paired with adeno-associated virus (AAV)-mediated delivery of a CCR5-targeting donor template to achieve highly efficient targeted recombination in primary human T cells. This method consistently achieved 8 to 60% rates of homology-directed recombination into the CCR5 locus in T cells, with over 80% of cells modified with an MND-GFP expression cassette exhibiting biallelic modification. MND-GFP-modified T cells maintained a diverse repertoire and engrafted in immune-deficient mice as efficiently as unmodified cells. Using this method, we integrated sequences coding chimeric antigen receptors (CARs) into the CCR5 locus, and the resulting targeted CAR T cells exhibited antitumor or anti-HIV activity. Alternatively, we introduced the C46 HIV fusion inhibitor, generating T cell populations with high rates of biallelic CCR5 disruption paired with potential protection from HIV with CXCR4 co-receptor tropism. Finally, this protocol was applied to adult human mobilized CD34(+) cells, resulting in 15 to 20% homologous gene targeting. Our results demonstrate that high-efficiency targeted integration is feasible in primary human hematopoietic cells and highlight the potential of gene editing to engineer T cell products with myriad functional properties.


Subject(s)
Deoxyribonucleases/metabolism , Dependovirus/metabolism , Hematopoietic Stem Cells/metabolism , Receptors, CCR5/metabolism , Adult , Antigens, CD34/metabolism , CD3 Complex/metabolism , Cells, Cultured , DNA Repair , Genetic Loci , Genetic Therapy , Green Fluorescent Proteins/metabolism , Humans , RNA Editing/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
7.
Nucleic Acids Res ; 42(10): 6463-75, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24682825

ABSTRACT

LAGLIDADG homing endonucleases (LHEs) are compact endonucleases with 20-22 bp recognition sites, and thus are ideal scaffolds for engineering site-specific DNA cleavage enzymes for genome editing applications. Here, we describe a general approach to LHE engineering that combines rational design with directed evolution, using a yeast surface display high-throughput cleavage selection. This approach was employed to alter the binding and cleavage specificity of the I-Anil LHE to recognize a mutation in the mouse Bruton tyrosine kinase (Btk) gene causative for mouse X-linked immunodeficiency (XID)-a model of human X-linked agammaglobulinemia (XLA). The required re-targeting of I-AniI involved progressive resculpting of the DNA contact interface to accommodate nine base differences from the native cleavage sequence. The enzyme emerging from the progressive engineering process was specific for the XID mutant allele versus the wild-type (WT) allele, and exhibited activity equivalent to WT I-AniI in vitro and in cellulo reporter assays. Fusion of the enzyme to a site-specific DNA binding domain of transcription activator-like effector (TALE) resulted in a further enhancement of gene editing efficiency. These results illustrate the potential of LHE enzymes as specific and efficient tools for therapeutic genome engineering.


Subject(s)
Endodeoxyribonucleases/chemistry , Protein-Tyrosine Kinases/genetics , Agammaglobulinaemia Tyrosine Kinase , Animals , Cells, Cultured , DNA Cleavage , DNA-Binding Proteins/chemistry , Directed Molecular Evolution , Endodeoxyribonucleases/metabolism , Genetic Loci , Genomics , HEK293 Cells , Homologous Recombination , Humans , Indicators and Reagents , Mice , Mutation , Protein Structure, Tertiary
8.
Mol Ther ; 20(1): 204-13, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22031238

ABSTRACT

Osteogenesis imperfecta (OI) is caused by dominant mutations in the type I collagen genes. In principle, the skeletal abnormalities of OI could be treated by transplantation of patient-specific, bone-forming cells that no longer express the mutant gene. Here, we develop this approach by isolating mesenchymal cells from OI patients, inactivating their mutant collagen genes by adeno-associated virus (AAV)-mediated gene targeting, and deriving induced pluripotent stem cells (iPSCs) that were expanded and differentiated into mesenchymal stem cells (iMSCs). Gene-targeted iMSCs produced normal collagen and formed bone in vivo, but were less senescent and proliferated more than bone-derived MSCs. To generate iPSCs that would be more appropriate for clinical use, the reprogramming and selectable marker transgenes were removed by Cre recombinase. These results demonstrate that the combination of gene targeting and iPSC derivation can be used to produce potentially therapeutic cells from patients with genetic disease.


Subject(s)
Collagen/biosynthesis , Collagen/genetics , Genetic Therapy , Induced Pluripotent Stem Cells/transplantation , Osteogenesis Imperfecta/therapy , Osteogenesis/genetics , Adolescent , Cell Differentiation , Child , Child, Preschool , Gene Order , Gene Targeting/methods , Gene Transfer Techniques , Humans , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Osteogenesis Imperfecta/genetics , Transgenes
9.
Nat Protoc ; 6(4): 482-501, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21455185

ABSTRACT

Gene targeting with adeno-associated virus (AAV) vectors has been demonstrated in multiple human cell types, with targeting frequencies ranging from 10(-5) to 10(-2) per infected cell. These targeting frequencies are 1-4 logs higher than those obtained by conventional transfection or electroporation approaches. A wide variety of different types of mutations can be introduced into chromosomal loci with high fidelity and without genotoxicity. Here we provide a detailed protocol for gene targeting in human cells with AAV vectors. We describe methods for vector design, stock preparation and titration. Optimized transduction protocols are provided for human pluripotent stem cells, mesenchymal stem cells, fibroblasts and transformed cell lines, as well as a method for identifying targeted clones by Southern blots. This protocol (from vector design through a single round of targeting and screening) can be completed in ∼10 weeks; each subsequent round of targeting and screening should take an additional 7 weeks.


Subject(s)
Dependovirus/genetics , Gene Targeting/methods , Animals , Cells, Cultured , Genetic Vectors , Humans , Mice , Transduction, Genetic/methods
10.
Mol Ther ; 18(6): 1192-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20407427

ABSTRACT

Precise genetic manipulation of human pluripotent stem cells will be required to realize their scientific and therapeutic potential. Here, we show that adeno-associated virus (AAV) gene targeting vectors can be used to genetically engineer human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Different types of sequence-specific changes, including the creation and correction of mutations, were introduced into the human HPRT1 and HMGA1 genes (HPRT1 mutations being responsible for Lesch-Nyhan syndrome). Gene targeting occurred at high frequencies in both ESCs and iPSCs, with over 1% of all colony-forming units (CFUs) undergoing targeting in some experiments. AAV vectors could also be used to target genes in human fibroblasts that were subsequently used to derive iPSCs. Accurate and efficient targeting took place with minimal or no cytotoxicity, and most of the gene-targeted stem cells produced were euploid and pluripotent.


Subject(s)
Dependovirus/genetics , Gene Targeting , Genetic Engineering , Pluripotent Stem Cells , Humans , Hypoxanthine Phosphoribosyltransferase/genetics
11.
Nat Med ; 14(1): 93-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18157138

ABSTRACT

Recent successes in treating genetic immunodeficiencies have demonstrated the therapeutic potential of stem cell gene therapy. However, the use of gammaretroviral vectors in these trials led to insertional activation of nearby oncogenes and leukemias in some study subjects, prompting studies of modified or alternative vector systems. Here we describe the use of foamy virus vectors to treat canine leukocyte adhesion deficiency (CLAD). Four of five dogs with CLAD that received nonmyeloablative conditioning and infusion of autologous, CD34+ hematopoietic stem cells transduced by a foamy virus vector expressing canine CD18 had complete reversal of the CLAD phenotype, which was sustained more than 2 years after infusion. In vitro assays showed correction of the lymphocyte proliferation and neutrophil adhesion defects that characterize CLAD. There were no genotoxic complications, and integration site analysis showed polyclonality of transduced cells and a decreased risk of integration near oncogenes as compared to gammaretroviral vectors. These results represent the first successful use of a foamy virus vector to treat a genetic disease, to our knowledge, and suggest that foamy virus vectors will be effective in treating human hematopoietic diseases.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , Leukocyte-Adhesion Deficiency Syndrome/therapy , Leukocytes/cytology , Spumavirus/genetics , Animals , Antigens, CD34/biosynthesis , Bone Marrow Cells/metabolism , Cell Adhesion , Cell Proliferation , Dogs , Hematopoietic Stem Cells/metabolism , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/veterinary , Lymphocytes/metabolism , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL