Abstract

Context: Chronic lymphoproliferative disorder of natural killer cells (CLPD-NK) is a subset of large granular lymphocyte (LGL) leukemia included as a provisional entity in the 2016 WHO classification of lymphoid neoplasms, which is characterized by clonal expansion of NK cells with no standardized therapy. STAT3 and TET2 mutations were previously each reported in 15%–30% of cases; however, the genomic basis of the remaining cases is still unknown. Objective: We aimed to perform a comprehensive genomic and transcriptomic analysis of CLPD-NK to elucidate the genomic landscape, investigate mechanisms of pathogenesis, and improve diagnostic classification and treatment strategies. Methods: CLPD-NK samples (n=121) were examined by whole-genome, targeted, and/or whole-transcriptome sequencing. Structural modeling, G-protein signaling, β-arrestin signaling, G-protein-coupled receptor (GPCR) for CCL22, CCR4 internalization, and chemotaxis were examined. Cell line-derived xenografts were used to examine cell proliferation and phenotype. Results: Somatic mutations in the chemokine CCL22 were found in 21.5% of CLPD-NK patients but not in 2837 cases of other hematological malignancies, including T-LGL and other NK leukemias. Clinically, there was a trend to an association with cutaneous complications and/or neurological symptoms. CCL22 mutations were enriched at the highly conserved residues Leu45, Pro46, and Pro79 and were mutually exclusive of STAT3 mutations. CCL22 mutations were associated with dysregulated gene expression, similar to normal CD56bright NK cells, with enrichment for activation of cytokine signaling pathways, representing a distinct subgroup. Most CCL22 mutations were substitutions to Arg, a basic residue, and Leu45Arg was predicted to promote electrostatic interactions with CCR4-Tyr22/Glu23 acidic residues. The mutations resulted in ligand-biased CCR4 signaling, in which activation of G-protein signaling was unchanged, but recruitment of β-arrestin, which mediates receptor internalization, was impaired, which, in turn, resulted in enhanced cell chemotaxis. CCL22 mutations drove enhanced cell proliferation in vivo in hIL-15-transgenic mice and induced an expression profile resembling that of primary samples, indicating the importance of microenvironmental crosstalk in pathogenesis. Conclusions: Somatic CCL22 mutations defined a new subgroup of CLPD-NK similar to CD56bright normal NK cells and represented a novel mechanism of leukemogenesis in which gain-of-function chemokine mutations drive tumorigenesis by biased GPCR signaling and dysregulation of microenvironmental crosstalk.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call