Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
JCI Insight ; 9(10)2024 May 22.
Article in English | MEDLINE | ID: mdl-38775150

ABSTRACT

This study lays the groundwork for future lentivirus-mediated gene therapy in patients with Diamond Blackfan anemia (DBA) caused by mutations in ribosomal protein S19 (RPS19), showing evidence of a new safe and effective therapy. The data show that, unlike patients with Fanconi anemia (FA), the hematopoietic stem cell (HSC) reservoir of patients with DBA was not significantly reduced, suggesting that collection of these cells should not constitute a remarkable restriction for DBA gene therapy. Subsequently, 2 clinically applicable lentiviral vectors were developed. In the former lentiviral vector, PGK.CoRPS19 LV, a codon-optimized version of RPS19 was driven by the phosphoglycerate kinase promoter (PGK) already used in different gene therapy trials, including FA gene therapy. In the latter one, EF1α.CoRPS19 LV, RPS19 expression was driven by the elongation factor alpha short promoter, EF1α(s). Preclinical experiments showed that transduction of DBA patient CD34+ cells with the PGK.CoRPS19 LV restored erythroid differentiation, and demonstrated the long-term repopulating properties of corrected DBA CD34+ cells, providing evidence of improved erythroid maturation. Concomitantly, long-term restoration of ribosomal biogenesis was verified using a potentially novel method applicable to patients' blood cells, based on ribosomal RNA methylation analyses. Finally, in vivo safety studies and proviral insertion site analyses showed that lentivirus-mediated gene therapy was nontoxic.


Subject(s)
Anemia, Diamond-Blackfan , Genetic Therapy , Genetic Vectors , Hematopoietic Stem Cells , Lentivirus , Ribosomal Proteins , Anemia, Diamond-Blackfan/therapy , Anemia, Diamond-Blackfan/genetics , Humans , Genetic Therapy/methods , Lentivirus/genetics , Ribosomal Proteins/genetics , Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Animals , Mice , Male , Female , Ribosomes/metabolism , Ribosomes/genetics , Promoter Regions, Genetic , Mutation , Hematopoietic Stem Cell Transplantation/methods
2.
Cytotherapy ; 25(3): 277-285, 2023 03.
Article in English | MEDLINE | ID: mdl-36610813

ABSTRACT

The clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system has revolutionized the gene editing field, making it possible to interrupt, insert or replace a sequence of interest with high precision in the human genome. Its easy design and wide applicability open up a variety of therapeutic alternatives for the treatment of genetic diseases. Indeed, very promising approaches for the correction of hematological disorders have been developed in the recent years, based on the self-renewal and multipotent differentiation properties of hematopoietic stem and progenitor cells, which make this cell subset the ideal target for gene therapy purposes. This technology has been applied in different congenital blood disorders, such as primary immunodeficiencies, X-linked severe combined immunodeficiency, X-linked chronic granulomatous disease or Wiskott-Aldrich syndrome, and inherited bone marrow failure syndromes, such as Fanconi anemia, congenital amegakaryocytic thrombocytopenia or severe congenital neutropenia. Furthermore, CRISPR/Cas9-based gene editing has been implemented successfully as a novel therapy for cancer immunotherapy, by the development of promising strategies such as the use of oncolytic viruses or adoptive cellular therapy to the chimeric antigen receptor-T-cell therapy. Therefore, considering the variety of genes and mutations affected, we can take advantage of the different DNA repair mechanisms by CRISPR/Cas9 in different manners, from homology-directed repair to non-homologous-end-joining to the latest emerging technologies such as base and prime editing. Although the delivery systems into hematopoietic stem and progenitor cells are still the bottleneck of this technology, some of the advances in genome editing shown in this review have already reached a clinical stage and show very promising preliminary results.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Humans , Gene Editing/methods , CRISPR-Cas Systems/genetics , Genetic Therapy/methods , Mutation
3.
Nat Commun ; 13(1): 6900, 2022 11 12.
Article in English | MEDLINE | ID: mdl-36371486

ABSTRACT

Fanconi Anemia (FA) is a debilitating genetic disorder with a wide range of severe symptoms including bone marrow failure and predisposition to cancer. CRISPR-Cas genome editing manipulates genotypes by harnessing DNA repair and has been proposed as a potential cure for FA. But FA is caused by deficiencies in DNA repair itself, preventing the use of editing strategies such as homology directed repair. Recently developed base editing (BE) systems do not rely on double stranded DNA breaks and might be used to target mutations in FA genes, but this remains to be tested. Here we develop a proof of concept therapeutic base editing strategy to address two of the most prevalent FANCA mutations in patient hematopoietic stem and progenitor cells. We find that optimizing adenine base editor construct, vector type, guide RNA format, and delivery conditions leads to very effective genetic modification in multiple FA patient backgrounds. Optimized base editing restored FANCA expression, molecular function of the FA pathway, and phenotypic resistance to crosslinking agents. ABE8e mediated editing in primary hematopoietic stem and progenitor cells from FA patients was both genotypically effective and restored FA pathway function, indicating the potential of base editing strategies for future clinical application in FA.


Subject(s)
Fanconi Anemia , Humans , Fanconi Anemia/genetics , Fanconi Anemia/therapy , Fanconi Anemia/metabolism , Adenine/metabolism , Hematopoietic Stem Cells/metabolism , Gene Editing , DNA Repair
4.
Mol Ther ; 29(6): 2008-2018, 2021 06 02.
Article in English | MEDLINE | ID: mdl-33609734

ABSTRACT

Genome-editing technologies that enable the introduction of precise changes in DNA sequences have the potential to lead to a new class of treatments for genetic diseases. Epidermolysis bullosa (EB) is a group of rare genetic disorders characterized by extreme skin fragility. The recessive dystrophic subtype of EB (RDEB), which has one of the most severe phenotypes, is caused by mutations in COL7A1. In this study, we report a gene-editing approach for ex vivo homology-directed repair (HDR)-based gene correction that uses the CRISPR-Cas9 system delivered as a ribonucleoprotein (RNP) complex in combination with donor DNA templates delivered by adeno-associated viral vectors (AAVs). We demonstrate sufficient mutation correction frequencies to achieve therapeutic benefit in primary RDEB keratinocytes containing different COL7A1 mutations as well as efficient HDR-mediated COL7A1 modification in healthy cord blood-derived CD34+ cells and mesenchymal stem cells (MSCs). These results are a proof of concept for HDR-mediated gene correction in different cell types with therapeutic potential for RDEB.


Subject(s)
Epidermolysis Bullosa Dystrophica/genetics , Gene Editing/methods , Genes, Recessive , Genetic Therapy/methods , Mutation , Recombinational DNA Repair , CRISPR-Cas Systems , Cell Line , Collagen Type VII/genetics , Dependovirus/genetics , Epidermolysis Bullosa Dystrophica/therapy , Gene Expression , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Keratinocytes/metabolism
5.
Cell Stem Cell ; 25(5): 607-621.e7, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31543367

ABSTRACT

Non-homologous end-joining (NHEJ) is the preferred mechanism used by hematopoietic stem cells (HSCs) to repair double-stranded DNA breaks and is particularly increased in cells deficient in the Fanconi anemia (FA) pathway. Here, we show feasible correction of compromised functional phenotypes in hematopoietic cells from multiple FA complementation groups, including FA-A, FA-C, FA-D1, and FA-D2. NHEJ-mediated repair of targeted CRISPR-Cas9-induced DNA breaks generated compensatory insertions and deletions that restore the coding frame of the mutated gene. NHEJ-mediated editing efficacy was initially verified in FA lymphoblastic cell lines and then in primary FA patient-derived CD34+ cells, which showed marked proliferative advantage and phenotypic correction both in vitro and after transplantation. Importantly, and in contrast to homologous directed repair, NHEJ efficiently targeted primitive human HSCs, indicating that NHEJ editing approaches may constitute a sound alternative for editing self-renewing human HSCs and consequently for treatment of FA and other monogenic diseases affecting the hematopoietic system.


Subject(s)
CRISPR-Cas Systems/genetics , DNA End-Joining Repair/genetics , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia/therapy , Gene Editing/methods , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation , Alleles , Animals , Antigens, CD34/metabolism , Cell Line , Cell Proliferation/genetics , DNA Breaks, Double-Stranded , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein/metabolism , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , High-Throughput Nucleotide Sequencing , Humans , INDEL Mutation , Mice , Mice, Inbred NOD , Mice, Nude
6.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...