Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Nat Genet ; 55(7): 1210-1220, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37400614

RESUMEN

Inducing fetal hemoglobin (HbF) in red blood cells can alleviate ß-thalassemia and sickle cell disease. We compared five strategies in CD34+ hematopoietic stem and progenitor cells, using either Cas9 nuclease or adenine base editors. The most potent modification was adenine base editor generation of γ-globin -175A>G. Homozygous -175A>G edited erythroid colonies expressed 81 ± 7% HbF versus 17 ± 11% in unedited controls, whereas HbF levels were lower and more variable for two Cas9 strategies targeting a BCL11A binding motif in the γ-globin promoter or a BCL11A erythroid enhancer. The -175A>G base edit also induced HbF more potently than a Cas9 approach in red blood cells generated after transplantation of CD34+ hematopoietic stem and progenitor cells into mice. Our data suggest a strategy for potent, uniform induction of HbF and provide insights into γ-globin gene regulation. More generally, we demonstrate that diverse indels generated by Cas9 can cause unexpected phenotypic variation that can be circumvented by base editing.


Asunto(s)
Anemia de Células Falciformes , Talasemia beta , Ratones , Animales , gamma-Globinas/genética , gamma-Globinas/metabolismo , Edición Génica , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Anemia de Células Falciformes/genética , Antígenos CD34/metabolismo , Talasemia beta/genética
3.
Nat Genet ; 54(9): 1417-1426, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35941187

RESUMEN

The fetal-to-adult switch in hemoglobin production is a model of developmental gene control with relevance to the treatment of hemoglobinopathies. The expression of transcription factor BCL11A, which represses fetal ß-type globin (HBG) genes in adult erythroid cells, is predominantly controlled at the transcriptional level but the underlying mechanism is unclear. We identify HIC2 as a repressor of BCL11A transcription. HIC2 and BCL11A are reciprocally expressed during development. Forced expression of HIC2 in adult erythroid cells inhibits BCL11A transcription and induces HBG expression. HIC2 binds to erythroid BCL11A enhancers to reduce chromatin accessibility and binding of transcription factor GATA1, diminishing enhancer activity and enhancer-promoter contacts. DNA-binding and crystallography studies reveal direct steric hindrance as one mechanism by which HIC2 inhibits GATA1 binding at a critical BCL11A enhancer. Conversely, loss of HIC2 in fetal erythroblasts increases enhancer accessibility, GATA1 binding and BCL11A transcription. HIC2 emerges as an evolutionarily conserved regulator of hemoglobin switching via developmental control of BCL11A.


Asunto(s)
Hemoglobinas , Factores de Transcripción de Tipo Kruppel , Proteínas Represoras , Proteínas Supresoras de Tumor , Proteínas Portadoras/genética , Células Eritroides/metabolismo , Hemoglobinas/genética , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Globinas beta/genética , Globinas beta/metabolismo , gamma-Globinas/genética
4.
Nat Genet ; 53(8): 1177-1186, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34341563

RESUMEN

Hereditary persistence of fetal hemoglobin (HPFH) ameliorates ß-hemoglobinopathies by inhibiting the developmental switch from γ-globin (HBG1/HBG2) to ß-globin (HBB) gene expression. Some forms of HPFH are associated with γ-globin promoter variants that either disrupt binding motifs for transcriptional repressors or create new motifs for transcriptional activators. How these variants sustain γ-globin gene expression postnatally remains undefined. We mapped γ-globin promoter sequences functionally in erythroid cells harboring different HPFH variants. Those that disrupt a BCL11A repressor binding element induce γ-globin expression by facilitating the recruitment of nuclear transcription factor Y (NF-Y) to a nearby proximal CCAAT box and GATA1 to an upstream motif. The proximal CCAAT element becomes dispensable for HPFH variants that generate new binding motifs for activators NF-Y or KLF1, but GATA1 recruitment remains essential. Our findings define distinct mechanisms through which transcription factors and their cis-regulatory elements activate γ-globin expression in different forms of HPFH, some of which are being recreated by therapeutic genome editing.


Asunto(s)
Factor de Unión a CCAAT/genética , Hemoglobina Fetal/genética , Factor de Transcripción GATA1/genética , gamma-Globinas/genética , Animales , Sitios de Unión , Células COS , Sistemas CRISPR-Cas , Línea Celular , Chlorocebus aethiops , Células Eritroides , Edición Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Humanos , Regiones Promotoras Genéticas , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...