Artificial mutations and natural variations in the CD46 molecules from human and monkey cells define regions important for measles virus binding

EC Hsu, RE Dörig, F Sarangi, A Marcil, C Iorio… - Journal of …, 1997 - Am Soc Microbiol
EC Hsu, RE Dörig, F Sarangi, A Marcil, C Iorio, CD Richardson
Journal of virology, 1997Am Soc Microbiol
CD46 was previously shown to be a primate-specific receptor for the Edmonston strain of
measles virus. This receptor consists of four short consensus regions (SCR1 to SCR4) which
normally function in complement regulation. Measles virus has recently been shown to
interact with SCR1 and SCR2. In this study, receptors on different types of monkey
erythrocytes were employed as" natural mutant proteins" to further define the virus binding
regions of CD46. Erythrocytes from African green monkeys and rhesus macaques …
CD46 was previously shown to be a primate-specific receptor for the Edmonston strain of measles virus. This receptor consists of four short consensus regions (SCR1 to SCR4) which normally function in complement regulation. Measles virus has recently been shown to interact with SCR1 and SCR2. In this study, receptors on different types of monkey erythrocytes were employed as "natural mutant proteins" to further define the virus binding regions of CD46. Erythrocytes from African green monkeys and rhesus macaques hemagglutinate in the presence of measles virus, while baboon erythrocytes were the least efficient of the Old World monkey cells used in these assays. Subsequent studies demonstrated that the SCR2 domain of baboon CD46 contained an Arg-to-Gln mutation at amino acid position 103 which accounted for reduced hemagglutination activity. Surprisingly, none of the New World monkey erythrocytes hemagglutinated in the presence of virus. Sequencing of cDNAs derived from the lymphocytes of these New World monkeys and analysis of their erythrocytes with SCR1-specific polyclonal antibodies indicated that the SCR1 domain was deleted in these cells. Additional experiments, which used 35 different site-specific mutations inserted into CD46, were performed to complement the preceding studies. The effects of these artificial mutations were documented with a convenient binding assay using insect cells expressing the measles virus hemagglutinin. Mutations which mimicked the change found in baboon CD46 or another which deleted the SCR2 glycosylation site reduced binding substantially. Another mutation which altered GluArg to AlaAla at positions 58 and 59, totally abolished binding. Finally, the epitopes for two monoclonal antibodies which inhibit measles virus attachment were mapped to the same regions implicated by mutagenesis.
American Society for Microbiology