-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

712 Ubiquitin Ligase SCF-FBXO11 Controls a Network of RNA-Binding Proteins and Splicing in MDS

Program: Oral and Poster Abstracts
Type: Oral
Session: 602. Myeloid Oncogenesis: Basic: RNA Splicing, Transcription, and Chromatin Dynamics
Hematology Disease Topics & Pathways:
Acute Myeloid Malignancies, Acquired Marrow Failure Syndromes, AML, Research, Fundamental Science, Bone Marrow Failure Syndromes, Diseases, Myeloid Malignancies, Biological Processes, molecular biology, pathogenesis
Monday, December 11, 2023: 11:15 AM

Madeline Niederkorn, PhD1, Lavanya Bezavada, MS2*, Anitria Cotton, MBA, BS3*, Lance Palmer, PhD1*, Trent Hall, PhD1*, Michael Schieber, MD, PhD4, Peng Xu, PhD5, Mitchell Weiss, MD, PhD1, Raajit K. Rampal, MD, PhD6 and John D. Crispino, PhD, MBA3

1St. Jude Children's Research Hospital, Memphis, TN
2Experimental Hematology, St. Jude Children's Research Hospital, Memphis, TN
3Division of Experimental Hematology, St Jude Children's Research Hospital, Memphis, TN
4Northwestern Medical Group, Lake Forest, IL
5Hematology Center, Cyrus Tang Medical Institute, Soochow University, Suzhou, Jiang Su, China
6Memorial Sloan Kettering Cancer Center, New York, NY

The F-Box protein FBXO11 is a substrate receptor of Skp-Cullin-F-Box (SCF) ubiquitin ligase complexes. Loss-of-function mutations in FBXO11 are associated with diffuse large B-cell lymphoma, and FBXO11 has been shown to regulate epigenetic drivers of erythroid cell maturation. In our prior study, we found that deletion of FBXO11 improved cell survival under cytokine deprivation in an MDS cell line. Furthermore, we had observed a striking decrease in FBXO11 protein levels in AML patient samples. However, how FBXO11 contributes to myeloid malignancies is unclear. Here, using RNA-sequencing, proteomics, and CRISPR-Cas9 screening, we reveal broad ubiquitylation of RNA-binding proteins (RBPs) and alternative splicing as a mechanism by which deregulation of FBXO11 enhances malignant cell function. In addition, we report the presence of mutations in the uncharacterized N-terminal extension of FBXO11 in both lymphoid and myeloid malignancies.

We first observed that heterozygous deletion of FBXO11 increases colony-forming ability of MDS-AML cell lines in vitro, while a 2-fold increase in expression results in decreased cell growth and apoptosis. In a transplant model with a retrovirally-driven Runx1 mutation, heterozygous deletion of Fbxo11 exacerbated neutropenia and shortened overall survival. However, depletion of FBXO11 in healthy human and murine HSPCs significantly impaired myeloid progenitor colony-formation, indicating a unique requirement for FBXO11 in myelopoiesis. Based on its tumor suppressive activity, we evaluated whole-exome sequencing data from patient samples of both lymphoid and myeloid malignancies for FBXO11 mutations. This analysis uncovered rare mutations in FBXO11 in myeloid malignancies and revealed a substantial cluster of previously unidentified mutations in the Q-rich, N-terminal extension of FBXO11. We found that this uncharacterized, extended protein isoform is predominantly expressed in both normal and malignant hematopoietic cells. In colony-forming assays, expression of the long isoform and several of the N-terminal mutations inhibited CD34+ progenitor function. On a cellular level, lack of this N-terminal extension in the short isoform resulted in aberrant subcellular localization of FBXO11 within the nucleus.

To elucidate the mechanism of FBXO11 in myeloid malignancy, we performed immunoprecipitation of FBXO11 followed by proteomics to identify endogenous FBXO11 complexes in MDS-AML cells. We integrated these data with ubiquitin proteomics of WT and FBXO11-KO MDS cells to identify candidate substrates of SCF-FBXO11-mediated ubiquitylation. FBXO11 complexes are significantly enriched for RNA-binding proteins affecting mRNA splicing, a process commonly deregulated in MDS. FBXO11-KO MDS cells displayed broad depletion of ubiquitylation in a network of RBPs. As expected, targeting FBXO11 did not affect the level of transcripts of interacting proteins; however, there were variable changes in total protein levels indicative of post-transcriptional regulation. We then applied a focused CRISPR-Cas9-KO screen of candidate SCF-FBXO11 substrates in MDS-AML cells depleted of FBXO11. We observed significant bi-directional effects in colony-forming ability mediated by several of the FBXO11-regulated RBPs including TRIM28, HNRNPU, NPM1, and SYNCRIP. Though FBXO11 is significantly decreased in MDS compared to healthy controls, we saw no difference in FBXO11 expression between splicing factor WT and mutant samples, suggesting that the function of FBXO11 in regulating mRNA splicing in MDS is independent of these mutations. Compared to healthy controls, FBXO11-low MDS samples exhibited a greater number of alternative splicing events, predominantly in skipped or mutually-exclusive exons. To experimentally test whether FBXO11 indeed affects RNA splicing, we utilized a bichromatic splicing reporter assay in MDS-AML cells which showed that depletion of FBXO11 is sufficient to alter splicing of the reading frame in the cassette exon, read out as a shift from GFP to RFP. Finally, STRING protein network analysis revealed that the alternative splicing events occurring in FBXO11-low MDS patient samples affect key nodes in protein translation and metabolism.

Collectively, our data support a model whereby the change in FBXO11-mediated ubiquitylation of RBPs drives alternative exon usage that exacerbates malignant cell function in MDS.

Disclosures: Weiss: GlaxoSmithKline: Consultancy; bluebird bio: Consultancy; Vertex Pharmaceuticals: Consultancy; Cellarity: Consultancy, Current equity holder in private company; Graphite Bio: Consultancy; Dyne: Consultancy; Novartis Inc.: Consultancy; Forma Therapeutics: Consultancy. Rampal: Zentalis: Research Funding; Kartos: Consultancy; Ryvu: Research Funding; Stemline: Research Funding; Constellation: Research Funding; Incyte: Research Funding; Dainippon: Consultancy; GSK-Sierra: Consultancy; Celgene-BMS: Consultancy; Morphosys/Constellation: Consultancy; Zentalis: Consultancy; Karyopharm: Consultancy; Servier: Consultancy; Sumitomo: Consultancy; Pharmaessentia: Consultancy; Galecto: Consultancy; CTI BioPharma Corp: Consultancy; Incyte: Consultancy. Crispino: Cellarity: Consultancy; SAB of Alethiomics: Other: Member.

*signifies non-member of ASH