Abstract

Hazara nairovirus (HAZV) is an enveloped trisegmented negative-strand RNA virus classified within the Nairoviridae family of the Bunyavirales order and a member of the same subtype as Crimean-Congo hemorrhagic fever virus, responsible for fatal human disease. Nairoviral subversion of cellular trafficking pathways to permit viral entry, gene expression, assembly, and egress is poorly understood. Here, we generated a recombinant HAZV expressing enhanced green fluorescent protein and used live-cell fluorescent imaging to screen an siRNA library targeting genes involved in cellular trafficking networks, the first such screen for a nairovirus. The screen revealed prominent roles for subunits of the coat protein 1 (COPI)-vesicle coatomer, which regulates retrograde trafficking of cargo between the Golgi apparatus and the endoplasmic reticulum, as well as intra-Golgi transport. We show the requirement of COPI-coatomer subunits impacted at least two stages of the HAZV replication cycle: an early stage prior to and including gene expression and also a later stage during assembly and egress of infectious virus, with COPI-knockdown reducing titers by approximately 1,000-fold. Treatment of HAZV-infected cells with brefeldin A (BFA), an inhibitor of Arf1 activation required for COPI coatomer formation, revealed that this late COPI-dependent stage was Arf1 dependent, consistent with the established role of Arf1 in COPI vesicle formation. In contrast, the early COPI-dependent stage was Arf1 independent, with neither BFA treatment nor siRNA-mediated ARF1 knockdown affecting HAZV gene expression. HAZV exploitation of COPI components in a noncanonical Arf1-independent process suggests that COPI coatomer components may perform roles unrelated to vesicle formation, adding further complexity to our understanding of cargo-mediated transport.IMPORTANCE Nairoviruses are tick-borne enveloped RNA viruses that include several pathogens responsible for fatal disease in humans and animals. Here, we analyzed host genes involved in trafficking networks to examine their involvement in nairovirus replication. We revealed important roles for genes that express multiple components of the COPI complex, which regulates transport of Golgi apparatus-resident cargos. COPI components influenced at least two stages of the nairovirus replication cycle: an early stage prior to and including gene expression and also a later stage during assembly of infectious virus, with COPI knockdown reducing titers by approximately 1,000-fold. Importantly, while the late stage was Arf1 dependent, as expected for canonical COPI vesicle formation, the early stage was found to be Arf1 independent, suggestive of a previously unreported function of COPI unrelated to vesicle formation. Collectively, these data improve our understanding of nairovirus host-pathogen interactions and suggest a new Arf1-independent role for components of the COPI coatomer complex.

Highlights

  • Hazara nairovirus (HAZV) is an enveloped trisegmented negativestrand RNA virus classified within the Nairoviridae family of the Bunyavirales order and a member of the same subtype as Crimean-Congo hemorrhagic fever virus, responsible for fatal human disease

  • brefeldin A (BFA) treatment markedly reduced the production of infectious HAZV, with a BFA concentration of 50 ng/ml reducing released titers by Ͼ100-fold. These results show that the HAZV replication cycle can be divided into at least two distinct phases based on the relative influence of coat protein 1 (COPI) and dependence on ADP-ribosylation factor 1 (Arf1) functionality: an early phase prior to and including protein production that exhibits a minor COPI dependence and is Arf1 independent, followed by a second phase involved in infectious virus production that is highly COPI dependent and dependent on Arf1 function

  • Here, we describe the development of rHAZV-eGFP, a modified recombinant HAZV

Read more

Summary

Introduction

Hazara nairovirus (HAZV) is an enveloped trisegmented negativestrand RNA virus classified within the Nairoviridae family of the Bunyavirales order and a member of the same subtype as Crimean-Congo hemorrhagic fever virus, responsible for fatal human disease. By analogy with Bunyamwera virus (BUNV), the prototypical Bunyavirales order member, one candidate location for these factories is the Golgi apparatus; BUNV establishes its replication factories surrounding the Golgi stacks [18], inducing the formation of specialized viral tubes that accumulate viral components, with subsequent assembly and maturation of virions involving the passage through Golgi subcompartments prior to virus budding from secretory vesicles [19, 20] This utilization of cellular endomembranes to support various viral processes is a common strategy adopted by enveloped viruses but is poorly understood for nairoviruses

Methods
Results
Conclusion
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