Your browser doesn't support javascript.
loading
let-7 miRNAs repress HIC2 to regulate BCL11A transcription and hemoglobin switching.
Huang, Peng; Peslak, Scott A; Shehu, Vanessa; Keller, Cheryl A; Giardine, Belinda; Shi, Junwei; Hardison, Ross C; Blobel, Gerd A; Khandros, Eugene.
Afiliación
  • Huang P; GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gyn
  • Peslak SA; Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
  • Shehu V; Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA.
  • Keller CA; Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA.
  • Giardine B; Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA.
  • Shi J; Genomics Research Incubator, Pennsylvania State University, University Park, PA.
  • Hardison RC; Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA.
  • Blobel GA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
  • Khandros E; Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA.
Blood ; 143(19): 1980-1991, 2024 May 09.
Article en En | MEDLINE | ID: mdl-38364109
ABSTRACT
ABSTRACT The switch from fetal hemoglobin (γ-globin, HBG) to adult hemoglobin (ß-globin, HBB) gene transcription in erythroid cells serves as a paradigm for a complex and clinically relevant developmental gene regulatory program. We previously identified HIC2 as a regulator of the switch by inhibiting the transcription of BCL11A, a key repressor of HBG production. HIC2 is highly expressed in fetal cells, but the mechanism of its regulation is unclear. Here we report that HIC2 developmental expression is controlled by microRNAs (miRNAs), as loss of global miRNA biogenesis through DICER1 depletion leads to upregulation of HIC2 and HBG messenger RNA. We identified the adult-expressed let-7 miRNA family as a direct posttranscriptional regulator of HIC2. Ectopic expression of let-7 in fetal cells lowered HIC2 levels, whereas inhibition of let-7 in adult erythroblasts increased HIC2 production, culminating in decommissioning of a BCL11A erythroid enhancer and reduced BCL11A transcription. HIC2 depletion in let-7-inhibited cells restored BCL11A-mediated repression of HBG. Together, these data establish that fetal hemoglobin silencing in adult erythroid cells is under the control of a miRNA-mediated inhibitory pathway (let-7 ⊣ HIC2 ⊣ BCL11A ⊣ HBG).
Asunto(s)

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Proteínas Represoras / Hemoglobina Fetal / MicroARNs / Factores de Transcripción de Tipo Kruppel Límite: Humans Idioma: En Revista: Blood Año: 2024 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Proteínas Represoras / Hemoglobina Fetal / MicroARNs / Factores de Transcripción de Tipo Kruppel Límite: Humans Idioma: En Revista: Blood Año: 2024 Tipo del documento: Article