Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 6 de 6
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Nat Commun ; 12(1): 4922, 2021 08 13.
Article de Anglais | MEDLINE | ID: mdl-34389729

RÉSUMÉ

CRISPR-Cas9 is a promising technology for gene therapy. However, the ON-target genotoxicity of CRISPR-Cas9 nuclease due to DNA double-strand breaks has received little attention and is probably underestimated. Here we report that genome editing targeting globin genes induces megabase-scale losses of heterozygosity (LOH) from the globin CRISPR-Cas9 cut-site to the telomere (5.2 Mb). In established lines, CRISPR-Cas9 nuclease induces frequent terminal chromosome 11p truncations and rare copy-neutral LOH. In primary hematopoietic progenitor/stem cells, we detect 1.1% of clones (7/648) with acquired megabase LOH induced by CRISPR-Cas9. In-depth analysis by SNP-array reveals the presence of copy-neutral LOH. This leads to 11p15.5 partial uniparental disomy, comprising two Chr11p15.5 imprinting centers (H19/IGF2:IG-DMR/IC1 and KCNQ1OT1:TSS-DMR/IC2) and impacting H19 and IGF2 expression. While this genotoxicity is a safety concern for CRISPR clinical trials, it is also an opportunity to model copy-neutral-LOH for genetic diseases and cancers.


Sujet(s)
Systèmes CRISPR-Cas , Édition de gène/méthodes , Globines/génétique , Cellules souches hématopoïétiques/métabolisme , Perte d'hétérozygotie/génétique , Délétion de séquence , Cellules cultivées , Délétion de segment de chromosome , Chromosomes humains de la paire 11/génétique , Méthylation de l'ADN , Expression des gènes , Cellules HEK293 , Cellules souches hématopoïétiques/cytologie , Humains , Facteur de croissance IGF-II/génétique , Polymorphisme de nucléotide simple , ARN long non codant/génétique
2.
Biochem Biophys Res Commun ; 569: 23-28, 2021 09 10.
Article de Anglais | MEDLINE | ID: mdl-34216994

RÉSUMÉ

Intravenous injections of human hematopoietic stem cells (hHSCs) is routinely used in clinic and for modeling hematopoiesis in mice. However, unspecific dilution in vascular system and non-hematopoietic organs challenges engraftment efficiency. Although spleen is capable of extra medullar hematopoiesis, its ability to support human HSC transplantation has never been evaluated. We demonstrate that intra-splenic injection results in high and sustained engraftment of hHSCs into immune-deficient mice, with higher chimerisms than with intravenous or intra-femoral injections. Our results support that spleen microenvironment provides a niche for HSCs amplification and offers a new route for efficient HSC transplantation.


Sujet(s)
Survie du greffon/physiologie , Transplantation de cellules souches hématopoïétiques/méthodes , Cellules souches hématopoïétiques/cytologie , Rate/cytologie , Animaux , Antigènes CD34/métabolisme , Femelle , Cytométrie en flux/méthodes , Hématopoïèse/physiologie , Cellules souches hématopoïétiques/métabolisme , Humains , Injections , Luciferases/génétique , Luciferases/métabolisme , Mesures de luminescence/méthodes , Souris de lignée NOD , Souris knockout , Souris SCID , Rate/métabolisme , Chimère obtenue par transplantation , Transplantation hétérologue
3.
Leukemia ; 31(1): 65-74, 2017 01.
Article de Anglais | MEDLINE | ID: mdl-27220663

RÉSUMÉ

Although tyrosine kinase inhibitors (TKIs) efficiently cure chronic myeloid leukemia (CML), they can fail to eradicate CML stem cells (CML-SCs). The mechanisms responsible for CML-SC survival need to be understood for designing therapies. Several previous studies suggest that TKIs could modulate CML-SC quiescence. Unfortunately, CML-SCs are insufficiently available. Induced pluripotent stem cells (iPSCs) offer a promising alternative. In this work, we used iPSCs derived from CML patients (Ph+). Ph+ iPSC clones expressed lower levels of stemness markers than normal iPSCs. BCR-ABL1 was found to be involved in stemness regulation and ERK1/2 to have a key role in the signaling pathway. TKIs unexpectedly promoted stemness marker expression in Ph+ iPSC clones. Imatinib also retained quiescence and induced stemness gene expression in CML-SCs. Our results suggest that TKIs might have a role in residual disease and confirm the need for a targeted therapy different from TKIs that could overcome the stemness-promoting effect caused by TKIs. Interestingly, a similar pro-stemness effect was observed in normal iPSCs and hematopoietic SCs. These findings could help to explain CML resistance mechanisms and the teratogenic side-effects of TKIs in embryonic cells.


Sujet(s)
Cellules souches pluripotentes induites/anatomopathologie , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Cellules souches tumorales/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Protéines de fusion bcr-abl/physiologie , Humains , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Système de signalisation des MAP kinases/physiologie , Cellules souches tumorales/anatomopathologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protein-tyrosine kinases/antagonistes et inhibiteurs , Cellules cancéreuses en culture
4.
Genomics ; 87(1): 84-92, 2006 Jan.
Article de Anglais | MEDLINE | ID: mdl-16314073

RÉSUMÉ

Congenital erythropoietic porphyria (CEP) is a recessive autosomal disorder characterized by a deficiency in uroporphyrinogen III synthase (UROS), the fourth enzyme of the heme biosynthetic pathway. The severity of the disease, the lack of specific treatment except for allogeneic bone marrow transplantation, and the knowledge of the molecular lesions are strong arguments for gene therapy. An animal model of CEP has been designed to evaluate the feasibility of retroviral gene transfer in hematopoietic stem cells. We have previously demonstrated that the knockout of the Uros gene is lethal in mice (Uros(del) model). This work describes the achievement of a knock-in model, which reproduces a mutation of the UROS gene responsible for a severe UROS deficiency in humans (P248Q missense mutant). Homozygous mice display erythrodontia, moderate photosensitivity, hepatosplenomegaly, and hemolytic anemia. Uroporphyrin (99% type I isomer) accumulates in urine. Total porphyrins are increased in erythrocytes and feces, while Uros enzymatic activity is below 1% of the normal level in the different tissues analyzed. These pathological findings closely mimic the CEP disease in humans and demonstrate that the Uros(mut248) mouse represents a suitable model of the human disease for pathophysiological, pharmaceutical, and therapeutic purposes.


Sujet(s)
Substitution d'acide aminé , Mutation faux-sens , Porphyrie érythropoïétique/enzymologie , Uroporphyrinogen III synthetase/génétique , Animaux , Transplantation de moelle osseuse , Modèles animaux de maladie humaine , Thérapie génétique , Souris , Souris transgéniques , Porphyrie érythropoïétique/anatomopathologie , Porphyrie érythropoïétique/thérapie , Uroporphyrinogen III synthetase/métabolisme , Uroporphyrines/métabolisme
5.
J Mol Med (Berl) ; 81(5): 310-20, 2003 May.
Article de Anglais | MEDLINE | ID: mdl-12721665

RÉSUMÉ

Congenital erythropoietic porphyria (CEP) is an inherited disease due to a deficiency in the uroporphyrinogen III synthase, the fourth enzyme of the heme biosynthesis pathway. It is characterized by accumulation of uroporphyrin I in the bone marrow, peripheral blood and other organs. The prognosis of CEP is poor, with death often occurring early in adult life. For severe transfusion-dependent cases, when allogeneic cell transplantation cannot be performed, the autografting of genetically modified primitive/stem cells may be the only alternative. In vitro gene transfer experiments have documented the feasibility of gene therapy via hematopoietic cells to treat this disease. In the present study lentiviral transduction of porphyric cell lines and primary CD34(+) cells with the therapeutic human uroporphyrinogen III synthase (UROS) cDNA resulted in both enzymatic and metabolic correction, as demonstrated by the increase in UROS activity and the suppression of porphyrin accumulation in transduced cells. Very high gene transfer efficiency (up to 90%) was achieved in both cell lines and CD34(+) cells without any selection. Expression of the transgene remained stable over long-term liquid culture. Furthermore, gene expression was maintained during in vitro erythroid differentiation of CD34(+) cells. Therefore the use of lentiviral vectors is promising for the future treatment of CEP patients by gene therapy.


Sujet(s)
Thérapie génétique , Lentivirus/génétique , Porphyrie érythropoïétique/thérapie , Uroporphyrinogen III synthetase/génétique , Adulte , Techniques de culture cellulaire , Différenciation cellulaire , Érythroblastes/métabolisme , Fluorescence , Expression des gènes , Vecteurs génétiques , Humains , Phénotype , Porphyrie érythropoïétique/génétique , Transduction génétique , Réplication virale
6.
Mol Ther ; 3(3): 411-7, 2001 Mar.
Article de Anglais | MEDLINE | ID: mdl-11273784

RÉSUMÉ

Congenital erythropoietic porphyria (CEP) is an inherited disease due to a deficiency in the uroporphyrinogen III synthase (UROS), the fourth enzyme of the heme pathway. It is characterized by accumulation of uroporphyrin I in the bone marrow, peripheral blood, and other organs. The onset of most cases occurs in infancy and the main symptoms are cutaneous photosensitivity and hemolysis. For severe transfusion-dependent cases, when allogeneic cell transplantation cannot be performed, autografting of genetically modified primitive/stem cells is the only alternative. In the present study, efficient mobilization of peripheral blood primitive CD34(+) cells was performed on a young adult CEP patient. Retroviral transduction of this cell population with the therapeutic human UROS (hUS) gene resulted in both enzymatic and metabolic correction of CD34(+)-derived cells, as demonstrated by the increase in UROS activity and by a 53% drop in porphyrin accumulation. A 10-24% gene transfer efficiency was achieved in the most primitive cells, as demonstrated by the expression of enhanced green fluorescent protein (EGFP) in long-term culture-initiating cells (LTC-IC). Furthermore, gene expression remained stable during in vitro erythroid differentiation. Therefore, these results are promising for the future treatment of CEP patients by gene therapy.


Sujet(s)
Antigènes CD34/métabolisme , Thérapie génétique , Cellules souches hématopoïétiques/métabolisme , Porphyrie érythropoïétique/thérapie , Retroviridae/génétique , Uroporphyrinogen III synthetase/génétique , Antigènes CD34/génétique , Moelle osseuse/enzymologie , Expression des gènes , Techniques de transfert de gènes , Vecteurs génétiques , Humains , Lentivirus/génétique , Porphyrines/métabolisme , Transduction génétique , Cellules cancéreuses en culture
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE