Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(11)2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37298481

RESUMEN

Beta-hemoglobinopathies are the most common genetic disorders worldwide, caused by a wide spectrum of mutations in the ß-globin locus, and associated with morbidity and early mortality in case of patient non-adherence to supportive treatment. Allogeneic transplantation of hematopoietic stem cells (allo-HSCT) used to be the only curative option, although the indispensable need for an HLA-matched donor markedly restricted its universal application. The evolution of gene therapy approaches made possible the ex vivo delivery of a therapeutic ß- or γ- globin gene into patient-derived hematopoietic stem cells followed by the transplantation of corrected cells into myeloablated patients, having led to high rates of transfusion independence (thalassemia) or complete resolution of painful crises (sickle cell disease-SCD). Hereditary persistence of fetal hemoglobin (HPFH), a syndrome characterized by increased γ-globin levels, when co-inherited with ß-thalassemia or SCD, converts hemoglobinopathies to a benign condition with mild clinical phenotype. The rapid development of precise genome editing tools (ZFN, TALENs, CRISPR/Cas9) over the last decade has allowed the targeted introduction of mutations, resulting in disease-modifying outcomes. In this context, genome editing tools have successfully been used for the introduction of HPFH-like mutations both in HBG1/HBG2 promoters or/and in the erythroid enhancer of BCL11A to increase HbF expression as an alternative curative approach for ß-hemoglobinopathies. The current investigation of new HbF modulators, such as ZBTB7A, KLF-1, SOX6, and ZNF410, further expands the range of possible genome editing targets. Importantly, genome editing approaches have recently reached clinical translation in trials investigating HbF reactivation in both SCD and thalassemic patients. Showing promising outcomes, these approaches are yet to be confirmed in long-term follow-up studies.


Asunto(s)
Anemia de Células Falciformes , Hemoglobinopatías , Talasemia beta , Humanos , Sistemas CRISPR-Cas , Línea Celular Tumoral , Factores de Transcripción/metabolismo , Proteínas de Unión al ADN/metabolismo , Hemoglobinopatías/genética , Hemoglobinopatías/terapia , Edición Génica/métodos , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Anemia de Células Falciformes/metabolismo , gamma-Globinas/genética , gamma-Globinas/metabolismo , Talasemia beta/genética , Talasemia beta/terapia , Talasemia beta/metabolismo
2.
Blood ; 141(17): 2085-2099, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-36800642

RESUMEN

Sickle cell disease (SCD) is a monogenic disease caused by a nucleotide mutation in the ß-globin gene. Current gene therapy studies are mainly focused on lentiviral vector-mediated gene addition or CRISPR/Cas9-mediated fetal globin reactivation, leaving the root cause unfixed. We developed a vectorized prime editing system that can directly repair the SCD mutation in hematopoietic stem cells (HSCs) in vivo in a SCD mouse model (CD46/Townes mice). Our approach involved a single intravenous injection of a nonintegrating, prime editor-expressing viral vector into mobilized CD46/Townes mice and low-dose drug selection in vivo. This procedure resulted in the correction of ∼40% of ßS alleles in HSCs. On average, 43% of sickle hemoglobin was replaced by adult hemoglobin, thereby greatly mitigating the SCD phenotypes. Transplantation in secondary recipients demonstrated that long-term repopulating HSCs were edited. Highly efficient target site editing was achieved with minimal generation of insertions and deletions and no detectable off-target editing. Because of its simplicity and portability, our in vivo prime editing approach has the potential for application in resource-poor countries where SCD is prevalent.


Asunto(s)
Anemia de Células Falciformes , Edición Génica , Ratones , Animales , Edición Génica/métodos , Sistemas CRISPR-Cas , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Células Madre Hematopoyéticas , Hemoglobina Falciforme/genética
3.
Genes (Basel) ; 13(12)2022 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-36553489

RESUMEN

The tremendous evolution of genome-editing tools in the last two decades has provided innovative and effective approaches for gene therapy of congenital and acquired diseases. Zinc-finger nucleases (ZFNs), transcription activator- like effector nucleases (TALENs) and CRISPR-Cas9 have been already applied by ex vivo hematopoietic stem cell (HSC) gene therapy in genetic diseases (i.e., Hemoglobinopathies, Fanconi anemia and hereditary Immunodeficiencies) as well as infectious diseases (i.e., HIV), and the recent development of CRISPR-Cas9-based systems using base and prime editors as well as epigenome editors has provided safer tools for gene therapy. The ex vivo approach for gene addition or editing of HSCs, however, is complex, invasive, technically challenging, costly and not free of toxicity. In vivo gene addition or editing promise to transform gene therapy from a highly sophisticated strategy to a "user-friendly' approach to eventually become a broadly available, highly accessible and potentially affordable treatment modality. In the present review article, based on the lessons gained by more than 3 decades of ex vivo HSC gene therapy, we discuss the concept, the tools, the progress made and the challenges to clinical translation of in vivo HSC gene editing.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Sistemas CRISPR-Cas/genética , Células Madre Hematopoyéticas , Terapia Genética , Nucleasas de los Efectores Tipo Activadores de la Transcripción
4.
JCI Insight ; 7(19)2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36006707

RESUMEN

Individuals with ß-thalassemia or sickle cell disease and hereditary persistence of fetal hemoglobin (HPFH) possessing 30% fetal hemoglobin (HbF) appear to be symptom free. Here, we used a nonintegrating HDAd5/35++ vector expressing a highly efficient and accurate version of an adenine base editor (ABE8e) to install, in vivo, a -113 A>G HPFH mutation in the γ-globin promoters in healthy CD46/ß-YAC mice carrying the human ß-globin locus. Our in vivo hematopoietic stem cell (HSC) editing/selection strategy involves only s.c. and i.v. injections and does not require myeloablation and HSC transplantation. In vivo HSC base editing in CD46/ß-YAC mice resulted in > 60% -113 A>G conversion, with 30% γ-globin of ß-globin expressed in 70% of erythrocytes. Importantly, no off-target editing at sites predicted by CIRCLE-Seq or in silico was detected. Furthermore, no critical alterations in the transcriptome of in vivo edited mice were found by RNA-Seq. In vitro, in HSCs from ß-thalassemia and patients with sickle cell disease, transduction with the base editor vector mediated efficient -113 A>G conversion and reactivation of γ-globin expression with subsequent phenotypic correction of erythroid cells. Because our in vivo base editing strategy is safe and technically simple, it has the potential for clinical application in developing countries where hemoglobinopathies are prevalent.


Asunto(s)
Anemia de Células Falciformes , Hemoglobinopatías , Talasemia beta , Adenina , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Animales , Sistemas CRISPR-Cas , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Edición Génica/métodos , Hemoglobinopatías/genética , Hemoglobinopatías/terapia , Humanos , Ratones , Globinas beta/genética , Talasemia beta/genética , Talasemia beta/terapia , gamma-Globinas/genética
5.
Heliyon ; 8(7): e09863, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35815135

RESUMEN

Background: A robust efficiency of mRNA vaccines against coronavirus disease-2019 has been demonstrated, however, the intended long-term protection against SARS-CoV-2 has been challenged by the waning humoral and cellular immunity over time, leading to a third vaccination dose recommendation for immunocompetent individuals, six months after completion of primary mRNA vaccination. Methods: We here measured humoral responses via an immunoassay measuring SARS-CoV-2 neutralizing antibodies and T-cell responses using Elispot for interferon-γ 1- and 8- months post full BNT162b2 vaccination, in 10 health-care professionals. To explore whether the declining abundance of coronavirus-specific T-cells (CoV-2-STs) truly reflects decreased capacity for viral control, rather than the attenuating viral stimulus over time, we modeled ex vivo the T-cellular response upon viral challenge in fully vaccinated immunocompetent individuals, 1- and 8-months post BNT162b2. Findings: Notwithstanding the declining CoV-2-neutralizing antibodies and CoV-2-STs, re-challenged CoV-2-STs, 1- and 8-months post vaccination, presented similar functional characteristics including high cytotoxicity against both the unmutated virus and the delta variant. Interpretation: These findings suggest robust and sustained cellular immune response upon SARS-CοV-2 antigen exposure, 8 months post mRNA vaccination, despite declining CοV-2-STs over time in the presence of an attenuating viral stimulus.

6.
Blood ; 138(17): 1540-1553, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34086867

RESUMEN

Thalassemia or sickle cell patients with hereditary persistence of fetal hemoglobin (HbF) have an ameliorated clinical phenotype and, in some cases, can achieve transfusion independence. Inactivation via genome editing of γ-globin developmental suppressors, such as BCL11A or LRF/ZBTB7A, or of their binding sites, have been shown to significantly increase expression of endogenous HbF. To broaden the therapeutic window beyond a single-editing approach, we have explored combinations of cis- and trans-editing targets to enhance HbF reactivation. Multiplex mutagenesis in adult CD34+ cells was well tolerated and did not lead to any detectable defect in the cells' proliferation and differentiation, either in vitro or in vivo. The combination of 1 trans and 1 cis mutation resulted in high editing retention in vivo, coupled with almost pancellular HbF expression in NBSGW mice. The greater in vivo performance of this combination was also recapitulated using a novel helper-dependent adenoviral-CRISPR vector (HD-Ad-dualCRISPR) in CD34+ cells from ß-thalassemia patients transplanted to NBSGW mice. A pronounced increase in HbF expression was observed in human red blood cells in mice with established predominant ß0/ß0-thalassemic hemopoiesis after in vivo injection of the HD-Ad-dualCRISPR vector. Collectively, our data suggest that the combination of cis and trans fetal globin reactivation mutations has the potential to significantly increase HbF both totally and on a per cell basis over single editing and could thus provide significant clinical benefit to patients with severe ß-globin phenotype.


Asunto(s)
Antígenos CD34/genética , Hemoglobina Fetal/genética , Mutagénesis , Talasemia beta/genética , Adulto , Animales , Sistemas CRISPR-Cas , Células Cultivadas , Edición Génica , Terapia Genética , Humanos , Ratones , Talasemia beta/terapia , gamma-Globinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...