Conserved Oligomeric Golgi (COG) Complex Proteins Facilitate Orthopoxvirus Entry, Fusion and Spread

Although orthopoxviruses (OPXV) are known to encode a majority of the genes required for replication in host cells, genome-wide genetic screens have revealed that several host pathways are indispensable for OPXV infection. Through a haploid genetic screen, we previously identified several host genes required for monkeypox virus (MPXV) infection, including the individual genes that form the conserved oligomeric Golgi (COG) complex. The COG complex is an eight-protein (COG1–COG8) vesicle tethering complex important for regulating membrane trafficking, glycosylation enzymes, and maintaining Golgi structure. In this study, we investigated the role of the COG complex in OPXV infection using cell lines with individual COG gene knockout (KO) mutations. COG KO cells infected with MPXV and vaccinia virus (VACV) produced small plaques and a lower virus yield compared to wild type (WT) cells. In cells where the KO phenotype was reversed using a rescue plasmid, the size of virus plaques increased demonstrating a direct link between the decrease in viral spread and the KO of COG genes. KO cells infected with VACV displayed lower levels of viral fusion and entry compared to WT suggesting that the COG complex is important for early events in OPXV infection. Additionally, fewer actin tails were observed in VACV-infected KO cells compared to WT. Since COG complex proteins are required for cellular trafficking of glycosylated membrane proteins, the disruption of this process due to lack of individual COG complex proteins may potentially impair the virus-cell interactions required for viral entry and egress. These data validate that the COG complex previously identified in our genetic screens plays a role in OPXV infection.

[1]  T. Brummelkamp,et al.  A Genome-Wide Haploid Genetic Screen Identifies Heparan Sulfate-Associated Genes and the Macropinocytosis Modulator TMED10 as Factors Supporting Vaccinia Virus Infection , 2019, Journal of Virology.

[2]  V. Lupashin,et al.  Membrane detachment is not essential for COG complex function , 2018, Molecular biology of the cell.

[3]  Jennifer A Cundiff,et al.  A Brucella Type IV Effector Targets the COG Tethering Complex to Remodel Host Secretory Traffic and Promote Intracellular Replication. , 2017, Cell host & microbe.

[4]  J. Carette,et al.  Monkeypox Virus Host Factor Screen Using Haploid Cells Identifies Essential Role of GARP Complex in Extracellular Virus Formation , 2017, Journal of Virology.

[5]  Tali Pechenick Jowers,et al.  Vaccinia Virus Uses Retromer-Independent Cellular Retrograde Transport Pathways To Facilitate the Wrapping of Intracellular Mature Virions during Virus Morphogenesis , 2016, Journal of Virology.

[6]  B. Moss,et al.  Retrograde Transport from Early Endosomes to the trans-Golgi Network Enables Membrane Wrapping and Egress of Vaccinia Virus Virions , 2016, Journal of Virology.

[7]  D. Ungar,et al.  COG Complex Complexities: Detailed Characterization of a Complete Set of HEK293T Cells Lacking Individual COG Subunits , 2016, Front. Cell Dev. Biol..

[8]  V. Lupashin,et al.  Creating Knockouts of Conserved Oligomeric Golgi Complex Subunits Using CRISPR-Mediated Gene Editing Paired with a Selection Strategy Based on Glycosylation Defects Associated with Impaired COG Complex Function. , 2016, Methods in molecular biology.

[9]  D. Dykxhoorn,et al.  Target silencing of components of the conserved oligomeric Golgi complex impairs HIV-1 replication. , 2014, Virus research.

[10]  Orland R. Gonzalez,et al.  A Loss of Function Analysis of Host Factors Influencing Vaccinia virus Replication by RNA Interference , 2014, PloS one.

[11]  Eugen C. Buehler,et al.  Human genome-wide RNAi screen reveals a role for nuclear pore proteins in poxvirus morphogenesis , 2013, Proceedings of the National Academy of Sciences.

[12]  B. Moss,et al.  Orthopoxvirus species and strain differences in cell entry , 2012, Virology.

[13]  V. Lupashin,et al.  Chlamydia trachomatis hijacks intra‐Golgi COG complex‐dependent vesicle trafficking pathway , 2012, Cellular microbiology.

[14]  B. Moss Poxvirus Cell Entry: How Many Proteins Does it Take? , 2012, Viruses.

[15]  B. Moss,et al.  The Membrane Fusion Step of Vaccinia Virus Entry Is Cooperatively Mediated by Multiple Viral Proteins and Host Cell Components , 2011, PLoS pathogens.

[16]  Richard D. Smith,et al.  Conserved oligomeric Golgi complex specifically regulates the maintenance of Golgi glycosylation machinery. , 2011, Glycobiology.

[17]  J. Bonifacino,et al.  Dual Roles of the Mammalian GARP Complex in Tethering and SNARE Complex Assembly at the trans-Golgi Network , 2009, Molecular and Cellular Biology.

[18]  M. Law,et al.  Acidic residues in the membrane-proximal stalk region of vaccinia virus protein B5 are required for glycosaminoglycan-mediated disruption of the extracellular enveloped virus outer membrane , 2009, The Journal of general virology.

[19]  B. Moss,et al.  Vaccinia virus strain differences in cell attachment and entry. , 2009, Virology.

[20]  Wen Chang,et al.  Vaccinia Virus 4c (A26L) Protein on Intracellular Mature Virus Binds to the Extracellular Cellular Matrix Laminin , 2006, Journal of Virology.

[21]  B. Moss,et al.  Vaccinia Virus Entry into Cells via a Low-pH-Dependent Endosomal Pathway , 2006, Journal of Virology.

[22]  M. Law,et al.  Ligand-induced and nonfusogenic dissolution of a viral membrane. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[23]  M. Krieger,et al.  Subunit Architecture of the Conserved Oligomeric Golgi Complex* , 2005, Journal of Biological Chemistry.

[24]  J. Krijnse Locker,et al.  Plasma Membrane Budding as an Alternative Release Mechanism of the Extracellular Enveloped Form of Vaccinia Virus from HeLa Cells , 2003, Journal of Virology.

[25]  R. Duden,et al.  The Sec34/Sec35p complex, a Ypt1p effector required for retrograde intra-Golgi trafficking, interacts with Golgi SNAREs and COPI vesicle coat proteins , 2002, The Journal of cell biology.

[26]  M. Law,et al.  Vaccinia virus utilizes microtubules for movement to the cell surface , 2001, The Journal of cell biology.

[27]  Geoffrey L. Smith,et al.  Vaccinia Virus F12L Protein Is Required for Actin Tail Formation, Normal Plaque Size, and Virulence , 2000, Journal of Virology.

[28]  Wen Chang,et al.  Vaccinia Virus Envelope H3L Protein Binds to Cell Surface Heparan Sulfate and Is Important for Intracellular Mature Virion Morphogenesis and Virus Infection In Vitro and In Vivo , 2000, Journal of Virology.

[29]  C. Chung,et al.  A27L Protein Mediates Vaccinia Virus Interaction with Cell Surface Heparan Sulfate , 1998, Journal of Virology.

[30]  M. Ericsson,et al.  Assembly of vaccinia virus: the second wrapping cisterna is derived from the trans Golgi network , 1994, Journal of virology.

[31]  J. Tooze,et al.  Progeny vaccinia and human cytomegalovirus particles utilize early endosomal cisternae for their envelopes. , 1993, European journal of cell biology.

[32]  K. Weber,et al.  Golgi-derived membranes that contain an acylated viral polypeptide are used for vaccinia virus envelopment , 1985, Journal of virology.

[33]  G. Stokes High-voltage electron microscope study of the release of vaccinia virus from whole cells , 1976, Journal of virology.