-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.

74 Blvrb Mutation Induces Thrombocytosis Via Redox Dysregulation in the Heme Degradation Pathway

Disorders of Platelet Number or Function
Program: Oral and Poster Abstracts
Type: Oral
Session: 311. Disorders of Platelet Number or Function: Genetics of Inherited and Acquired Thrombocytopenias
Saturday, December 5, 2015: 12:15 PM
W315, Level 3 (Orange County Convention Center)

Song Wu, PhD1*, Zongdong Li, PhD2, Dmitri V Gnatenko, PhD2*, BeiBei Zhang3*, Lu Zhao4*, Lisa Malone2*, Nedialka Markova, PhD5*, Timothy Mantle, PhD6*, Natasha Nesbitt, PhD5* and Wadie F. Bahou, MD2

1Department of Applied Mathematics & Statistics, Stony Brook Univeristy, Stony Brook, NY
2Department of Medicine, Stony Brook University, Stony Brook, NY
3Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY
4Department of Applied Mathematics & Statistics, Stony Brook University, Stony Brook, NY
5Department of Medicine, Stony Brook Univeristy, Stony Brook, NY
6Department of Biochemistry, Trinity College, Dublin 2, Ireland

Known genetic loci influencing blood cell production account for <10% of platelet and red blood cell variability, and thrombopoietin (Tpo)/c-MPL liganding is dispensable for definitive thrombopoiesis establishing that fundamentally important modifier loci remain unelucidated.  We completed RNASeq of highly-purified platelets from 7 essential thrombocythemia (ET) subjects [4 harbored the JAK2V617F mutation and 3 were genotypically normal] and 5 healthy controls, and developed an iterative algorithm to identify 33 non-synonymous SNVs that are putatively linked to the ET phenotype.  Transcripts possessing these candidate SNVs were enriched on average in early-stage megakaryocyte/erythroid progenitors (MEPs), and became more restricted in terminally-differentiating megakaryocytes and erythroblasts.   Genotypic studies using an expanded ET cohort (N = 36), followed by statistical association analyses using controls from the 1000 Human Genomes Project and an independently-genotyped cohort of healthy controls (N = 208), established that 5 SNVs (excluding JAK2V617F) were associated with ET.  A single mutation (BLVRBS111L; p = 0.0006) retained its significance as a thrombocytosis risk allele using genotypic data from a secondary cohort with reactive (non-clonal) thrombocytosis (RT, N = 53), suggesting a function as an independent driver mutation of enhanced thrombopoiesis. The BLVRB (biliverdin IXβ reductase) gene functions downstream of heme oxygenase(s)-1 (inducible HMOX1) and -2 (constitutive HMOX2) within the heme degradation pathway to catalyze reduction of biliverdin (BV) tetrapyrrole(s) as an intermediary redox substrate in Bilirubin (BR) generation. Bacterially-expressed and purified recombinant BLVRBS111L showed defective enzymatic activity [compared to BLVRBWT (wild-type)] using flavin mononucleotide [flavin reductase (FR) activity; p <0.0001] and BV IXβ dimethyl esters (biliverdin reductase (BVR) activity; p <0.0001), the latter specifically generated by coupled heme oxidation as verdin-restricted BLVRB activity probes.  The loss-of-mutation BLVRBS111L NAD(P)H-dependent redox coupling caused higher baseline ROS (reactive oxygen species) accumulation in lentivirus-transduced CD34+-derived induced pluripotent stem cells (iPSC/BLVRBS111L), results sharply contrasting with ROS neutralization in iPSC/BLVRBWT exposed to tert-butyl hydroperoxide (TBHP) as the oxidant stress source (p < 0.00001).  Disparate redox coupling/ROS handling in genetically-modified primary CD34+ multipotential progenitor cultures established disproportionate expansion of primitive CFU-GEMMs in CD34+/BLVRBS111L (p = 0.001), and an absolute increase of BFU-E in CD34+/BLVRBWT (p = 0.001).  Collagen-based progenitor cultures demonstrated a statistically-significant increase of CD41+ CFU-MKs in CD34+/BLVRBS111L cells (p <0.01) with no increased CFU-MKs in CD34+/BLVRBWT cells. Cumulative distribution plots of parallel Tpo-suspension cultures confirmed divergent ROS accumulation between MK/BLVRBWT and MK/BLVRBS111L, differences that were identifiable pre-terminal differentiation (Day 0, p = 0.0007), most pronounced at Day 5 corresponding to peak MK BLVRB expression across genotypes (p = 8 x 10-7), and persistent at terminal differentiation (Day 10; p = 0.05).  Maximally disparate ROS accumulation (Day 5) corresponded to greatest size disparity and temporally-earlier (and sustained at Day 10) CD41 expression in MK/BLVRBS111L (p = 0.03).  A bilineage (Tpo/Epo) differentiation model, designed to characterize erythroid/megakaryocyte (E/Meg) progenitor balance arising from common MEPs demonstrated no evidence for differential Mk (CD41+/Glycophorin A-) lineage balance, suggesting that BLVRBS111L ROS-promoting effects accelerate post-commitment expansion downstream of MEP lineage fate decisions.  These data provide the first evidence linking redox coupling to MK lineage fate and expansion in humans, either via activity as a flavin reductase, in partnered electron exchange with an unidentified protein, or as a verdin-regulated redox coupler regulated by heme degradation and isomer-restricted BV IXβ (or IXδ, IXγ) generation; in principle, development of BLVRB-specific redox inhibitors represent innovative approaches to selectively alter a regulatory pathway controlling MK lineage expansion and human platelet counts.

Disclosures: No relevant conflicts of interest to declare.

*signifies non-member of ASH