[HTML][HTML] Allelic polymorphism determines surface expression or intracellular retention of the human NK cell receptor KIR2DL5A (CD158f)

E Cisneros, E Estefanía, C Vilches - Frontiers in immunology, 2017 - frontiersin.org
E Cisneros, E Estefanía, C Vilches
Frontiers in immunology, 2017frontiersin.org
KIR2DL5 (CD158f) is the most recently identified inhibitory member of human killer-cell Ig-
like receptors (KIRs), which enable NK cells to sense self-HLA. Unlike KIR2DL1–3,
recognizing HLA-C allotypes through Ig-like domains of the D1–D2 type, KIR2DL5 shares a
D0–D2 configuration with KIR2DL4, and its ligands have not been identified. KIR2DL5 is
encoded by two paralogous genes displaying copy number variation and allelic
polymorphism—KIR2DL5A and KIR2DL5B. UP-R1 mAb, raised against the common allele …
KIR2DL5 (CD158f) is the most recently identified inhibitory member of human killer-cell Ig-like receptors (KIRs), which enable NK cells to sense self-HLA. Unlike KIR2DL1–3, recognizing HLA-C allotypes through Ig-like domains of the D1–D2 type, KIR2DL5 shares a D0–D2 configuration with KIR2DL4, and its ligands have not been identified. KIR2DL5 is encoded by two paralogous genes displaying copy number variation and allelic polymorphism—KIR2DL5A and KIR2DL5B. UP-R1 mAb, raised against the common allele KIR2DL5A*001, enables specific KIR2DL5 detection. However, not every KIR2DL5+ individual has NK cells staining with UP-R1, discrepancy explained in part by epigenetically silent KIR2DL5B alleles with a distinctive substitution in a promoter RUNX-binding site. Furthermore, we show here that the transcribed allele KIR2DL5A*005, second most common of its locus, fails to confer NK cells UP-R1 reactivity, phenotype explained by inefficacious transport of its product to the cell surface. Two amino acid substitutions distinguish the KIR2DL5A*005 and *001 coding regions. Western blot, flow cytometry, and confocal microscopy analyses of cells transfected with tagged constructs demonstrate that a serine substitution for glycine-174, conserved in most KIR, is mainly responsible for KIR2DL5A*005 intracellular retention, and it also affects mAb recognition. In contrast, substitution of aspartate for asparagine 152 has only a minor effect on surface expression, despite destroying an otherwise conserved N-glycosylation site. Our results help to explain the variable expression profile of KIR2DL5+ subjects and indicate that functional polymorphisms in both its promoter and its coding regions are critical for understanding the KIR2DL5 role in immunity and its importance for human health.
Frontiers