Your browser doesn't support javascript.
loading
HRI depletion cooperates with pharmacologic inducers to elevate fetal hemoglobin and reduce sickle cell formation.
Peslak, Scott A; Khandros, Eugene; Huang, Peng; Lan, Xianjiang; Geronimo, Carly L; Grevet, Jeremy D; Abdulmalik, Osheiza; Zhang, Zhe; Giardine, Belinda M; Keller, Cheryl A; Shi, Junwei; Hardison, Ross C; Blobel, Gerd A.
Afiliación
  • Peslak SA; Division of Hematology/Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA.
  • Khandros E; Division of Hematology and.
  • Huang P; Division of Hematology and.
  • Lan X; Division of Hematology and.
  • Geronimo CL; Division of Hematology and.
  • Grevet JD; Division of Hematology and.
  • Abdulmalik O; Division of Hematology and.
  • Zhang Z; Division of Hematology and.
  • Giardine BM; Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA.
  • Keller CA; Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA; and.
  • Shi J; Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA; and.
  • Hardison RC; Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
  • Blobel GA; Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, PA; and.
Blood Adv ; 4(18): 4560-4572, 2020 09 22.
Article en En | MEDLINE | ID: mdl-32956454
ABSTRACT
Increasing fetal hemoglobin (HbF) provides clinical benefit in patients with sickle cell disease (SCD). We recently identified heme-regulated inhibitor (HRI, EIF2AK1), as a novel HbF regulator. Because HRI is an erythroid-specific protein kinase, it presents a potential target for pharmacologic intervention. We found that maximal HbF induction required >80% to 85% HRI depletion. Because it remains unclear whether this degree of HRI inhibition can be achieved pharmacologically, we explored whether HRI knockdown can be combined with pharmacologic HbF inducers to achieve greater HbF production and minimize potential adverse effects associated with treatments. Strongly cooperative HbF induction was observed when HRI depletion was combined with exposure to pomalidomide or the EHMT1/2 inhibitor UNC0638, but not to hydroxyurea. Mechanistically, reduction in the levels of the HbF repressor BCL11A reflected the cooperativity of HRI loss and pomalidomide treatment, whereas UNC0638 did not modulate BCL11A levels. In conjunction with HRI loss, pomalidomide maintained its HbF-inducing activity at 10-fold lower concentrations, in which condition there were minimal observed detrimental effects on erythroid cell maturation and viability, as well as fewer alterations in the erythroid transcriptome. When tested in cells from patients with SCD, combining HRI depletion with pomalidomide or UNC0638 achieved up to 50% to 60% HbF and 45% to 50% HbF, respectively, as measured by high-performance liquid chromatography, and markedly counteracted cell sickling. In summary, this study provides a foundation for the exploration of combining future small-molecule HRI inhibitors with additional pharmacologic HbF inducers to maximize HbF production and preserve erythroid cell functionality for the treatment of SCD and other hemoglobinopathies.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Hemoglobina Fetal / Anemia de Células Falciformes Límite: Humans Idioma: En Revista: Blood Adv Año: 2020 Tipo del documento: Article País de afiliación: Panamá

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Hemoglobina Fetal / Anemia de Células Falciformes Límite: Humans Idioma: En Revista: Blood Adv Año: 2020 Tipo del documento: Article País de afiliación: Panamá