4.6 Article

Identification and characterization of RBM12 as a novel regulator of fetal hemoglobin expression

Journal

BLOOD ADVANCES
Volume 6, Issue 23, Pages 5956-5968

Publisher

ELSEVIER
DOI: 10.1182/bloodadvances.2022007904

Keywords

-

Categories

Funding

  1. National Institute of Diabetes and Digestive and Kidney Diseases [NIDDK] [DK106829]
  2. National Institutes of Health Hematopoiesis Training Grant [5T32DK007780-19]
  3. NIDDK [1F31DK122649, K08DK129716, K08DK128571, R24DK106766]
  4. Doris Duke Charitable Foundation Physician Scientist Fellowship Grant [2020062]
  5. National Heart, Lung, and Blood Institute (NHLBI) grant [R01HL119479]
  6. DiGaetano family

Ask authors/readers for more resources

This study identifies the RNA binding protein RBM12 as a novel suppressor of fetal hemoglobin (HbF) expression. Depletion of RBM12 induces HbF expression and attenuates cell sickling in erythroid cells derived from patients with sickle cell disease (SCD), with minimal detrimental effects on cell maturation.
The fetal-to-adult hemoglobin transition is clinically relevant because reactivation of fetal hemoglobin (HbF) significantly reduces morbidity and mortality associated with sickle cell disease (SCD) and ss-thalassemia. Most studies on the developmental regulation of the globin genes, including genome-wide genetics screens, have focused on DNA binding proteins, including BCL11A and ZBTB7A/LRF and their cofactors. Our understanding of RNA binding proteins (RBPs) in this process is much more limited. Two RBPs, LIN28B and IGF2BP1, are known posttranscriptional regulators of HbF production, but a global view of RBPs is still lacking. Here, we carried out a CRISPR/Cas9-based screen targeting RBPs harboring RNA methyltransferase and/or RNA recognition motif (RRM) domains and identified RNA binding motif 12 (RBM12) as a novel HbF suppressor. Depletion of RBM12 induced HbF expression and attenuated cell sickling in erythroid cells derived from patients with SCD with minimal detrimental effects on cell maturation. Transcriptome and proteome profiling revealed that RBM12 functions independently of major known HbF regulators. Enhanced cross-linking and immunoprecipitation followed by high-throughput sequencing revealed strong preferential binding of RBM12 to 5' untranslated regions of transcripts, narrowing down the mechanism of RBM12 action. Notably, we pinpointed the first of 5 RRM domains as essential, and, in conjunction with a linker domain, sufficient for RBM12-mediated HbF regulation. Our characterization of RBM12 as a negative regulator of HbF points to an additional regulatory layer of the fetal-to-adult hemoglobin switch and broadens the pool of potential therapeutic targets for SCD and ss-thalassemia.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available