4.6 Article

Cytidine Monophosphate N-Acetylneuraminic Acid Synthetase and Solute Carrier Family 35 Member A1 Are Required for Reovirus Binding and Infection

Journal

JOURNAL OF VIROLOGY
Volume 95, Issue 2, Pages -

Publisher

AMER SOC MICROBIOLOGY
DOI: 10.1128/JVI.01571-20

Keywords

solute carrier family 35 member A1; sialic acid; viral attachment; reovirus; microglia; cytidine monophosphate N-acetylneuraminic acid synthetase

Categories

Funding

  1. U.S. Public Health Service [R01 AI038296, R00 DK116666, K08 AI128043, U19 AI109725]
  2. Burroughs Welcome Fund Career Award for Medical Scientists
  3. Heinz Endowments

Ask authors/readers for more resources

Through a CRISPR cell survival screen, two genes - Cmas and Slc35a1 were identified as host genes required for reovirus infection of microglial cells by promoting sialic acid expression on the cell surface. These findings expand understanding of receptors engaged by reovirus on microglial cells and may lead to new strategies for targeting microglial cells for oncolytic applications.
Engagement of cell surface receptors by viruses is a critical determinant of viral tropism and disease. The reovirus attachment protein cr1 binds sialylated glycans and proteinaceous receptors to mediate infection, but the specific requirements for different cell types are not entirely known. To identify host factors required for reovirus-induced cell death, we conducted a CRISPR-knockout screen targeting over 20,000 genes in murine microglial BV2 cells. Candidate genes required for reovirus to cause cell death were highly enriched for sialic acid synthesis and transport. Two of the top candidates identified, CMP N-acetylneuraminic acid synthetase (Cmas) and solute carrier family 35 member A1 (Slc35a1), promote sialic acid expression on the cell surface. Two reovirus strains that differ in the capacity to bind sialic acid, T3SA(+) and T3SA(-), were used to evaluate Cmas and Slc35a1 as potential host genes required for reovirus infection. Following CRISPR-Cas9 disruption of either gene, cell surface expression of sialic acid was diminished. These results correlated with decreased binding of strain T3SA(+), which is capable of engaging sialic acid. Disruption of either gene did not alter the low-level binding of T3SA(-), which does not engage sialic acid. Furthermore, infectivity of T3SA(+) was diminished to levels similar to those of T3SA(-) in cells lacking Cmas and Slc35a1 by CRISPR ablation. However, exogenous expression of Cmas and Slc35a1 into the respective null cells restored sialic acid expression and T3SA(+) binding and infectivity. These results demonstrate that Cmas and Slc35a1, which mediate cell surface expression of sialic acid, are required in murine microglial cells for efficient reovirus binding and infection. IMPORTANCE Attachment factors and receptors are important determinants of dissemination and tropism during reovirus-induced disease. In a CRISPR cell survival screen, we discovered two genes, Cmas and Slc35a1, which encode proteins required for sialic acid expression on the cell surface and mediate reovirus infection of microglial cells. This work elucidates host genes that render microglial cells susceptible to reovirus infection and expands current understanding of the receptors on microglial cells that are engaged by reovirus. Such knowledge may lead to new strategies to selectively target microglial cells for oncolytic applications.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available