KaLwRij/C57Bl (KaLwRij, “Kal-ridge”) is a spontaneous mutant mouse strain predisposed to monoclonal gammopathy of uncertain significance (MGUS) and parallels many features of human pathology including progression to multiple myeloma (MM). In African Americans, an increased prevalence of MM is due to an increased risk of MGUS development (rather than increased rate of progression from MGUS) and is likely therefore mediated by germline MGUS risk alleles. In both humans and mice, the genetic basis for MGUS/MM risk is unknown. We sought to identify the genetic basis for MGUS/MM risk in KaLwRij mice to shed light on genes or pathways that might mediate risk in humans. We employed a novel integrative computational analysis, combining KaLwRij/B6 modified haplotype block analysis and low-power human MM/control single nucleotide polymorphism (SNP) data. We used high-density SNP arrays to establish genetic distances of KaLwRij from 11 another diverse inbred strains and confirmed close homology to parent C57Bl/6. Haplotype block analysis identified genetic loci that diverged from C57Bl/6 and included over 500 genes. We filtered the haplotype list using previously published RNA expression data from the bone marrow of C57Bl/6 and KaLwRij mice, to identify 71 differentially expressed genes. As a final filter, we used SNP data from a genome wide association study (GWAS) of human Caucasian MM case and normal volunteer samples (∼300 each) to identify SNPs differentially represented in MM patients. Combining these sets yielded four (4) candidate genes. Our top candidate, Samsn1 (Hacs1), is a negative regulator of B-cell activation and interestingly, was first identified as a gene differentially expressed in MM. Strikingly, we found that KaLwRij mice had a complete germline deletion of the Samsn1 gene. Samsn1 is expressed within the hematopoetic compartment, including B-cells, macrophages, and osteoclasts (OC), key cell types that participate in MM pathology. Consistent with the reported inhibitory function of SAMSN1, KaLwRij had enhanced naïve B-cell response and constitutive Lyn phosphorylation. Further, we found that KaLwRij macrophages and OC also have constitutive Lyn activation consistent activation of cytokine signaling pathways in stromal cells. Absence of Samsn1 may lead to unregulated B-cell and macrophage/OC activation, priming both a pro-tumor bone microenvironment and B-cells for development of MGUS and predisposition to MM. Our approach focused on germline loci rather than malignant cell genetic alterations, advancing our understanding of host predisposition. Identification of the gene or genes predisposing patients to MGUS/MM will provide novel targets for identifying at-risk individuals and will potentially allow the develop of preventative therapeutics for MM.

Disclosures:

No relevant conflicts of interest to declare.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution