1
|
Ogura T, Okada T. Observation of biological and emulsion samples by newly developed three-dimensional impedance scanning electron microscopy. Comput Struct Biotechnol J 2024; 23:4064-4076. [PMID: 39628906 PMCID: PMC11613192 DOI: 10.1016/j.csbj.2024.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 12/06/2024] Open
Abstract
Imaging at nanometre-scale resolution is indispensable for many scientific fields such as biology, chemistry, material science and nanotechnology. Scanning electron microscopes (SEM) are widely used as important tools for the nanometre-scale analysis of various samples. However, because of the vacuum inside the SEM, a typical analysis requires fixation of samples, a drying process, and staining with heavy metals. Therefore, there is a need for convenient and minimally invasive methods of observing samples in solution. Recently, we have developed a new type of impedance microscopy, multi-frequency impedance SEM (IP-SEM), which allows nanoscale imaging of various specimens in water with minimal radiation damage. Here, we report a new IP-SEM system equipped with a linear-array terminal, which allows eight tilted images to be observed in a single capture by applying eight frequencies of input signals to each electrode. Furthermore, we developed a three-dimensional (3D) reconstruction method based on the Simulated Annealing (SA) algorithm, which enables us to construct a high-precision 3D model from the 8 tilted images. The method reported here can be easily used for 3D structural analysis of various biological samples, organic materials, and nanoparticles.
Collapse
Affiliation(s)
- Toshihiko Ogura
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Tomoko Okada
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| |
Collapse
|
2
|
Pranclova V, Nedvedova L, Kotounova E, Hönig V, Dvorakova M, Davidkova M, Bily T, Vancova M, Ruzek D, Palus M. Unraveling the role of human microglia in tick-borne encephalitis virus infection: insights into neuroinflammation and viral pathogenesis. Microbes Infect 2024; 26:105383. [PMID: 38942136 DOI: 10.1016/j.micinf.2024.105383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/09/2024] [Accepted: 06/20/2024] [Indexed: 06/30/2024]
Abstract
Tick-borne encephalitis virus (TBEV) is a neurotropic orthoflavivirus responsible for severe infections of the central nervous system. Although neurons are predominantly targeted, specific involvement of microglia in pathogenesis of TBE is not yet fully understood. In this study, the susceptibility of human microglia to TBEV is investigated, focusing on productive infection and different immune responses of different viral strains. We investigated primary human microglia and two immortalized microglial cell lines exposed to three TBEV strains (Hypr, Neudörfl and 280), each differing in virulence. Our results show that all microglia cultures tested support long-term productive infections, regardless of the viral strain. In particular, immune response varied significantly with the viral strain, as shown by the differential secretion of cytokines and chemokines such as IP-10, MCP-1, IL-8 and IL-6, quantified using a Luminex 48-plex assay. The most virulent strain triggered the highest cytokine induction. Electron tomography revealed substantial ultrastructural changes in the infected microglia, despite the absence of cytopathic effects. These findings underscore the susceptibility of human microglia to TBEV and reveal strain-dependent variations in viral replication and immune responses, highlighting the complex role of microglia in TBEV-induced neuropathology and contribute to a deeper understanding of TBE pathogenesis and neuroinflammation.
Collapse
Affiliation(s)
- Veronika Pranclova
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Faculty of Science, University of South Bohemia, CZ-37005 Ceske Budejovice, Czech Republic
| | - Lenka Nedvedova
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Faculty of Science, University of South Bohemia, CZ-37005 Ceske Budejovice, Czech Republic
| | - Eliska Kotounova
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Faculty of Science, University of South Bohemia, CZ-37005 Ceske Budejovice, Czech Republic
| | - Vaclav Hönig
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Laboratory of Emerging Viral Infections, Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic
| | - Marketa Dvorakova
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic
| | - Marika Davidkova
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic
| | - Tomas Bily
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Faculty of Science, University of South Bohemia, CZ-37005 Ceske Budejovice, Czech Republic
| | - Marie Vancova
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Faculty of Science, University of South Bohemia, CZ-37005 Ceske Budejovice, Czech Republic
| | - Daniel Ruzek
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Laboratory of Emerging Viral Infections, Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, CZ-62500 Brno, Czech Republic
| | - Martin Palus
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Laboratory of Emerging Viral Infections, Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic.
| |
Collapse
|
3
|
Dahmane S, Schexnaydre E, Zhang J, Rosendal E, Chotiwan N, Kumari Singh B, Yau WL, Lundmark R, Barad B, Grotjahn DA, Liese S, Carlson A, Overby A, Carlson LA. Cryo-electron tomography reveals coupled flavivirus replication, budding and maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.618056. [PMID: 39416041 PMCID: PMC11482891 DOI: 10.1101/2024.10.13.618056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Flaviviruses replicate their genomes in replication organelles (ROs) formed as bud-like invaginations on the endoplasmic reticulum (ER) membrane, which also functions as the site for virion assembly. While this localization is well established, it is not known to what extent viral membrane remodeling, genome replication, virion assembly, and maturation are coordinated. Here, we imaged tick-borne flavivirus replication in human cells using cryo-electron tomography. We find that the RO membrane bud is shaped by a combination of a curvature-establishing coat and the pressure from intraluminal template RNA. A protein complex at the RO base extends to an adjacent membrane, where immature virions bud. Naturally occurring furin site variants determine whether virions mature in the immediate vicinity of ROs. We further visualize replication in mouse brain tissue by cryo-electron tomography. Taken together, these findings reveal a close spatial coupling of flavivirus genome replication, budding, and maturation.
Collapse
|
4
|
Ochwoto M, Offerdahl DK, Leung JM, Schwartz CL, Long D, Rosenke R, Stewart PE, Saturday GA, Bloom ME. Cytoarchitecture of ex vivo midgut cultures of unfed Ixodes scapularis infected with a tick-borne flavivirus. Ticks Tick Borne Dis 2024; 15:102301. [PMID: 38134511 PMCID: PMC10923016 DOI: 10.1016/j.ttbdis.2023.102301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023]
Abstract
A bite from an infected tick is the primary means of transmission for tick-borne flaviviruses (TBFV). Ticks ingest the virus while feeding on infected blood. The traditional view is that the virus first replicates in and transits the tick midgut prior to dissemination to other organs, including salivary glands. Thus, understanding TBFV infection in the tick midgut is a key first step in identifying potential countermeasures against infection. Ex vivo midgut cultures prepared from unfed adult female Ixodes scapularis ticks were viable and remained morphologically intact for more than 8 days. The midgut consisted of two clearly defined cell layers separated by a basement membrane: an exterior network of smooth muscle cells and an internal epithelium composed of digestive generative cells. The smooth muscle cells were arranged in a stellate circumferential pattern spaced at regular intervals along the long axis of midgut diverticula. When the cultures were infected with the TBFV Langat virus (LGTV), virus production increased by two logs with a peak at 96 hours post-infection. Infected cells were readily identified by immunofluorescence staining for the viral envelope protein, nonstructural protein 3 (NS3) and dsRNA. Microscopy of the stained cultures suggested that generative cells were the primary target for virus infection in the midgut. Infected cells exhibited an expansion of membranes derived from the endoplasmic reticulum; a finding consistent with TBFV infected cell cultures. Electron microscopy of infected cultures revealed virus particles in the basolateral region between epithelial cells. These results demonstrated LGTV replication in midgut generative cells of artificially infected, ex vivo cultures of unfed adult female I. scapularis ticks.
Collapse
Affiliation(s)
- Missiani Ochwoto
- Biology of Vector Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID, NIH, USA.
| | - Danielle K Offerdahl
- Biology of Vector Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID, NIH, USA
| | - Jacqueline M Leung
- Research Technologies Branch, Rocky Mountain Laboratories, NIAID, NIH, USA
| | - Cindi L Schwartz
- Research Technologies Branch, Rocky Mountain Laboratories, NIAID, NIH, USA
| | - Dan Long
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, NIAID, NIH, USA
| | - Rebecca Rosenke
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, NIAID, NIH, USA
| | - Philip E Stewart
- Biology of Vector Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID, NIH, USA
| | - Greg A Saturday
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, NIAID, NIH, USA
| | - Marshall E Bloom
- Biology of Vector Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID, NIH, USA.
| |
Collapse
|
5
|
Shiryaev SA, Cieplak P, Cheltsov A, Liddington RC, Terskikh AV. Dual function of Zika virus NS2B-NS3 protease. PLoS Pathog 2023; 19:e1011795. [PMID: 38011215 PMCID: PMC10723727 DOI: 10.1371/journal.ppat.1011795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/15/2023] [Accepted: 11/02/2023] [Indexed: 11/29/2023] Open
Abstract
Zika virus (ZIKV) serine protease, indispensable for viral polyprotein processing and replication, is composed of the membrane-anchored NS2B polypeptide and the N-terminal domain of the NS3 polypeptide (NS3pro). The C-terminal domain of the NS3 polypeptide (NS3hel) is necessary for helicase activity and contains an ATP-binding site. We discovered that ZIKV NS2B-NS3pro binds single-stranded RNA with a Kd of ~0.3 μM, suggesting a novel function. We tested various structural modifications of NS2B-NS3pro and observed that constructs stabilized in the recently discovered "super-open" conformation do not bind RNA. Likewise, stabilizing NS2B-NS3pro in the "closed" (proteolytically active) conformation using substrate inhibitors abolished RNA binding. We posit that RNA binding occurs when ZIKV NS2B-NS3pro adopts the "open" conformation, which we modeled using highly homologous dengue NS2B-NS3pro crystallized in the open conformation. We identified two positively charged fork-like structures present only in the open conformation of NS3pro. These forks are conserved across Flaviviridae family and could be aligned with the positively charged grove on NS3hel, providing a contiguous binding surface for the negative RNA strand exiting helicase. We propose a "reverse inchworm" model for a tightly intertwined NS2B-NS3 helicase-protease machinery, which suggests that NS2B-NS3pro cycles between open and super-open conformations to bind and release RNA enabling long-range NS3hel processivity. The transition to the closed conformation, likely induced by the substrate, enables the classical protease activity of NS2B-NS3pro.
Collapse
Affiliation(s)
- Sergey A. Shiryaev
- Sanford-Burnham-Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, La Jolla, California, United States of America
| | - Piotr Cieplak
- Sanford-Burnham-Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, La Jolla, California, United States of America
| | - Anton Cheltsov
- Q-mol LLC, San Diego, California, United States of America
| | - Robert C. Liddington
- Sanford-Burnham-Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, La Jolla, California, United States of America
| | - Alexey V. Terskikh
- Sanford-Burnham-Prebys Medical Discovery Institute, Infectious and Inflammatory Disease Center, La Jolla, California, United States of America
| |
Collapse
|
6
|
Perez-Valencia LJ, Vannella KM, Ramos-Benitez MJ, Sun J, Abu-Asab M, Dorward DW, Awad KS, Platt A, Jacobson E, Kindrachuk J, Chertow DS. Ebola virus shed glycoprotein is toxic to human T, B, and natural killer lymphocytes. iScience 2023; 26:107323. [PMID: 37529105 PMCID: PMC10387567 DOI: 10.1016/j.isci.2023.107323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 04/23/2023] [Accepted: 07/04/2023] [Indexed: 08/03/2023] Open
Abstract
Lymphocyte depletion is a distinctive feature of Ebola virus (EBOV) disease. The ectodomain of EBOV glycoprotein (GP) is cleaved off the surface of infected cells into circulation as shed GP. To test the hypothesis that shed GP induces lymphocyte death, we cultured primary human B, NK, or T cells with shed GP in vitro. We found that shed GP dependably decreased B, NK, and T cell viability across donors. B and NK cells exhibited higher susceptibility than T cells. Continuous monitoring revealed shed GP began to kill B and NK cells by 4 h and T cells by 5 h. We also demonstrated that shed GP-induced lymphocyte death can be both caspase dependent and caspase independent. Our data are evidence that the cytotoxic effect of shed GP on lymphocytes may contribute to EBOV disease and highlight the need for further research to clarify mechanisms of shed GP-induced death.
Collapse
Affiliation(s)
- Luis J. Perez-Valencia
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kevin M. Vannella
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marcos J. Ramos-Benitez
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mones Abu-Asab
- Section of Histopathology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David W. Dorward
- Microscopy Unit, Research Technology Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Keytam S. Awad
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew Platt
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eliana Jacobson
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jason Kindrachuk
- Laboratory of Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg MB R3E 0J9, Canada
| | - Daniel S. Chertow
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
7
|
Ratnayake OC, Chotiwan N, Saavedra-Rodriguez K, Perera R. The buzz in the field: the interaction between viruses, mosquitoes, and metabolism. Front Cell Infect Microbiol 2023; 13:1128577. [PMID: 37360524 PMCID: PMC10289420 DOI: 10.3389/fcimb.2023.1128577] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/24/2023] [Indexed: 06/28/2023] Open
Abstract
Among many medically important pathogens, arboviruses like dengue, Zika and chikungunya cause severe health and economic burdens especially in developing countries. These viruses are primarily vectored by mosquitoes. Having surmounted geographical barriers and threat of control strategies, these vectors continue to conquer many areas of the globe exposing more than half of the world's population to these viruses. Unfortunately, no medical interventions have been capable so far to produce successful vaccines or antivirals against many of these viruses. Thus, vector control remains the fundamental strategy to prevent disease transmission. The long-established understanding regarding the replication of these viruses is that they reshape both human and mosquito host cellular membranes upon infection for their replicative benefit. This leads to or is a result of significant alterations in lipid metabolism. Metabolism involves complex chemical reactions in the body that are essential for general physiological functions and survival of an organism. Finely tuned metabolic homeostases are maintained in healthy organisms. However, a simple stimulus like a viral infection can alter this homeostatic landscape driving considerable phenotypic change. Better comprehension of these mechanisms can serve as innovative control strategies against these vectors and viruses. Here, we review the metabolic basis of fundamental mosquito biology and virus-vector interactions. The cited work provides compelling evidence that targeting metabolism can be a paradigm shift and provide potent tools for vector control as well as tools to answer many unresolved questions and gaps in the field of arbovirology.
Collapse
Affiliation(s)
- Oshani C. Ratnayake
- Center for Vector-borne Infectious Diseases, Dept. of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Nunya Chotiwan
- Center for Vector-borne Infectious Diseases, Dept. of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Karla Saavedra-Rodriguez
- Center for Vector-borne Infectious Diseases, Dept. of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Rushika Perera
- Center for Vector-borne Infectious Diseases, Dept. of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
8
|
Scroggs SLP, Offerdahl DK, Stewart PE, Shaia C, Griffin AJ, Bloom ME. Of Murines and Humans: Modeling Persistent Powassan Disease in C57BL/6 Mice. mBio 2023; 14:e0360622. [PMID: 36809119 PMCID: PMC10128018 DOI: 10.1128/mbio.03606-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 01/09/2023] [Indexed: 02/23/2023] Open
Abstract
Powassan infection is caused by two closely related, tick-transmitted viruses of the genus Flavivirus (family Flaviviridae): Powassan virus lineage I (POWV) and lineage II (known as deer tick virus [DTV]). Infection is typically asymptomatic or mild but can progress to neuroinvasive disease. Approximately 10% of neuroinvasive cases are fatal, and half of the survivors experience long-term neurological sequelae. Understanding how these viruses cause long-term symptoms as well as the possible role of viral persistence is important for developing therapies. We intraperitoneally inoculated 6-week-old C57BL/6 mice (50% female) with 103 focus-forming units (FFU) DTV and assayed for infectious virus, viral RNA, and inflammation during acute infection and 21, 56, and 84 days postinfection (dpi). Although most mice (86%) were viremic 3 dpi, only 21% of the mice were symptomatic and 83% recovered. Infectious virus was detected only in the brains of mice sampled during the acute infection. Viral RNA was detected in the brain until 84 dpi, but the magnitude decreased over time. Meningitis and encephalitis were visible in acute mice and from mice sampled at 21 dpi. Inflammation was observed until 56 dpi in the brain and 84 dpi in the spinal cord, albeit at low levels. These results suggest that the long-term neurological symptoms associated with Powassan disease are likely caused by lingering viral RNA and chronic inflammation in the central nervous system rather than by a persistent, active viral infection. The C57BL/6 model of persistent Powassan mimics illness in humans and can be used to study the mechanisms of chronic disease. IMPORTANCE Half of Powassan infection survivors experience long-term, mild to severe neurological symptoms. The progression from acute to chronic Powassan disease is not well understood, severely limiting treatment and prevention options. Infection of C57BL/6 mice with DTV mimics clinical disease in humans, and the mice exhibit CNS inflammation and viral RNA persistence until at least 86 dpi, while infectious virus is undetectable after 12 dpi. These findings suggest that the long-term neurological symptoms of chronic Powassan disease are in part due the persistence of viral RNA and the corresponding long-term inflammation of the brain and spinal cord. Our work demonstrates that C57BL/6 mice can be used to study the pathogenesis of chronic Powassan disease.
Collapse
Affiliation(s)
- Stacey L. P. Scroggs
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
- Arthropod-Borne Animal Disease Research Unit, Center for Grain and Animal Health Research, Agricultural Research Service, United States Department of Agriculture, Manhattan, Kansas, USA
| | - Danielle K. Offerdahl
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Philip E. Stewart
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Amanda J. Griffin
- Office of the Chief, Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Marshall E. Bloom
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| |
Collapse
|
9
|
Goonawardane N, Upstone L, Harris M, Jones IM. Identification of Host Factors Differentially Induced by Clinically Diverse Strains of Tick-Borne Encephalitis Virus. J Virol 2022; 96:e0081822. [PMID: 36098513 PMCID: PMC9517736 DOI: 10.1128/jvi.00818-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/19/2022] [Indexed: 11/20/2022] Open
Abstract
Tick-borne encephalitis virus (TBEV) is an important human arthropod-borne virus that causes tick-borne encephalitis (TBE) in humans. TBEV acutely infects the central nervous system (CNS), leading to neurological symptoms of various severity. No therapeutics are currently available for TBEV-associated disease. Virus strains of various pathogenicity have been described, although the basis of their diverse clinical outcome remains undefined. Work with infectious TBEV requires high-level biocontainment, meaning model systems that can recapitulate the virus life cycle are highly sought. Here, we report the generation of a self-replicating, noninfectious TBEV replicon used to study properties of high (Hypr) and low (Vs) pathogenic TBEV isolates. Using a Spinach2 RNA aptamer and luciferase reporter system, we perform the first direct comparison of Hypr and Vs in cell culture. Infectious wild-type (WT) viruses and chimeras of the nonstructural proteins 3 (NS3) and 5 (NS5) were investigated in parallel to validate the replicon data. We show that Hypr replicates to higher levels than Vs in mammalian cells, but not in arthropod cells, and that the basis of these differences map to the NS5 region, encoding the methyltransferase and RNA polymerase. For both Hypr and Vs strains, NS5 and the viral genome localized to intracellular structures typical of positive-strand RNA viruses. Hypr was associated with significant activation of IRF-3, caspase-3, and caspase-8, while Vs activated Akt, affording protection against caspase-mediated apoptosis. Higher activation of stress-granule proteins TIAR and G3BPI were an additional early feature of Vs but not for Hypr. These findings highlight novel host cell responses driven by NS5 that may dictate the differential clinical characteristics of TBEV strains. This highlights the utility of the TBEV replicons for further virological characterization and antiviral drug screening. IMPORTANCE Tick-borne encephalitis virus (TBEV) is an emerging virus of the flavivirus family that is spread by ticks and causes neurological disease of various severity. No specific therapeutic treatments are available for TBE, and control in areas of endemicity is limited to vaccination. The pathology of TBEV ranges from mild to fatal, depending on the virus genotype. Characterization of TBEV isolates is challenging due to the requirement for high-containment facilities. Here, we described the construction of novel TBEV replicons that permit a molecular comparison of TBEV isolates of high and low pathogenicity.
Collapse
Affiliation(s)
- Niluka Goonawardane
- School of Biological Sciences, University of Reading, Reading, United Kingdom
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Laura Upstone
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Mark Harris
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Ian M. Jones
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|
10
|
O'Brien CA, Huang B, Warrilow D, Hazlewood JE, Bielefeldt-Ohmann H, Hall-Mendelin S, Pegg CL, Harrison JJ, Paramitha D, Newton ND, Schulz BL, Suhrbier A, Hobson-Peters J, Hall RA. Extended characterisation of five archival tick-borne viruses provides insights for virus discovery in Australian ticks. Parasit Vectors 2022; 15:59. [PMID: 35180893 PMCID: PMC8857802 DOI: 10.1186/s13071-022-05176-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/24/2022] [Indexed: 11/10/2022] Open
Abstract
Background A subset of Australians who have been bitten by ticks experience a complex of chronic and debilitating symptoms which cannot be attributed to the known pathogenic species of bacteria present in Australia. As a result, there has been a renewed effort to identify and characterise viruses in Australian terrestrial ticks. Recent transcriptome sequencing of Ixodes and Amblyomma ticks has revealed the presence of multiple virus sequences. However, without virus isolates our ability to understand the host range and pathogenesis of newly identified viruses is limited. We have established a successful method for high-throughput virus discovery and isolation in mosquitoes using antibodies to double-stranded RNA. In this study we sought to characterise five archival tick-borne viruses to adapt our virus discovery protocol for Australian ticks. Methods We performed virus characterisation using a combination of bioinformatic sequence analysis and in vitro techniques including replication kinetics, antigenic profiling, virus purification and mass spectrometry. Results Our sequence analysis of Nugget virus, Catch-me-Cave virus and Finch Creek virus revealed marked genetic stability in isolates collected from the same location approximately 30 years apart. We demonstrate that the Ixodes scapularis-derived ISE6 cell line supports replication of Australian members of the Flaviviridae, Nairoviridae, Phenuiviridae and Reoviridae families, including Saumarez Reef virus (SREV), a flavivirus isolated from the soft tick Ornithodoros capensis. While antibodies against double-stranded RNA could be used to detect replication of a tick-borne reovirus and mosquito-borne flavivirus, the tick-borne flaviviruses Gadgets Gully virus and SREV could not be detected using this method. Finally, four novel virus-like sequences were identified in transcriptome sequencing of the Australian native tick Ixodes holocyclus. Conclusions Genetic and antigenic characterisations of archival viruses in this study confirm that three viruses described in 2002 represent contemporary isolates of virus species first identified 30 years prior. Our findings with antibodies to double-stranded RNA highlight an unusual characteristic shared by two Australian tick-borne flaviviruses. Finally, comparative growth kinetics analyses of Australian tick-borne members of the Flaviviridae, Nairoviridae, Phenuiviridae and Reoviridae families in ISE6 and BSR cells will provide a useful resource for isolation of Australian tick-borne viruses using existing cell lines. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-022-05176-z.
Collapse
Affiliation(s)
- Caitlin A O'Brien
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, 4072, Australia.,Australian Infectious Disease Research Centre, GVN Center of Excellence, The University of Queensland and QIMR Berghofer Medical Research Institute, St Lucia, QLD, 4067, Australia
| | - Bixing Huang
- Public Health Virology, Forensic and Scientific Services, Department of Health, P.O. Box 594, Archerfield, QLD, Australia
| | - David Warrilow
- Public Health Virology, Forensic and Scientific Services, Department of Health, P.O. Box 594, Archerfield, QLD, Australia
| | - Jessamine E Hazlewood
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Helle Bielefeldt-Ohmann
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, 4072, Australia.,Australian Infectious Disease Research Centre, GVN Center of Excellence, The University of Queensland and QIMR Berghofer Medical Research Institute, St Lucia, QLD, 4067, Australia.,School of Veterinary Science, The University of Queensland, Gatton, QLD, Australia
| | - Sonja Hall-Mendelin
- Public Health Virology, Forensic and Scientific Services, Department of Health, P.O. Box 594, Archerfield, QLD, Australia
| | - Cassandra L Pegg
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, 4072, Australia.,Australian Infectious Disease Research Centre, GVN Center of Excellence, The University of Queensland and QIMR Berghofer Medical Research Institute, St Lucia, QLD, 4067, Australia
| | - Jessica J Harrison
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, 4072, Australia.,Australian Infectious Disease Research Centre, GVN Center of Excellence, The University of Queensland and QIMR Berghofer Medical Research Institute, St Lucia, QLD, 4067, Australia
| | - Devina Paramitha
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, 4072, Australia.,Australian Infectious Disease Research Centre, GVN Center of Excellence, The University of Queensland and QIMR Berghofer Medical Research Institute, St Lucia, QLD, 4067, Australia
| | - Natalee D Newton
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, 4072, Australia.,Australian Infectious Disease Research Centre, GVN Center of Excellence, The University of Queensland and QIMR Berghofer Medical Research Institute, St Lucia, QLD, 4067, Australia
| | - Benjamin L Schulz
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, 4072, Australia.,Australian Infectious Disease Research Centre, GVN Center of Excellence, The University of Queensland and QIMR Berghofer Medical Research Institute, St Lucia, QLD, 4067, Australia
| | - Andreas Suhrbier
- Australian Infectious Disease Research Centre, GVN Center of Excellence, The University of Queensland and QIMR Berghofer Medical Research Institute, St Lucia, QLD, 4067, Australia.,Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Jody Hobson-Peters
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, 4072, Australia.,Australian Infectious Disease Research Centre, GVN Center of Excellence, The University of Queensland and QIMR Berghofer Medical Research Institute, St Lucia, QLD, 4067, Australia
| | - Roy A Hall
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD, 4072, Australia. .,Australian Infectious Disease Research Centre, GVN Center of Excellence, The University of Queensland and QIMR Berghofer Medical Research Institute, St Lucia, QLD, 4067, Australia.
| |
Collapse
|
11
|
Differential characteristics of mammalian and tick-derived promoters to trigger protein expression in transfected tick cell lines. Ticks Tick Borne Dis 2022; 13:101906. [DOI: 10.1016/j.ttbdis.2022.101906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/05/2022] [Accepted: 01/14/2022] [Indexed: 11/19/2022]
|
12
|
Coronavirus RNA Synthesis Takes Place within Membrane-Bound Sites. Viruses 2021; 13:v13122540. [PMID: 34960809 PMCID: PMC8708976 DOI: 10.3390/v13122540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/01/2021] [Accepted: 12/15/2021] [Indexed: 12/22/2022] Open
Abstract
Infectious bronchitis virus (IBV), a gammacoronavirus, is an economically important virus to the poultry industry, as well as a significant welfare issue for chickens. As for all positive strand RNA viruses, IBV infection causes rearrangements of the host cell intracellular membranes to form replication organelles. Replication organelle formation is a highly conserved and vital step in the viral life cycle. Here, we investigate the localization of viral RNA synthesis and the link with replication organelles in host cells. We have shown that sites of viral RNA synthesis and virus-related dsRNA are associated with one another and, significantly, that they are located within a membrane-bound compartment within the cell. We have also shown that some viral RNA produced early in infection remains within these membranes throughout infection, while a proportion is trafficked to the cytoplasm. Importantly, we demonstrate conservation across all four coronavirus genera, including SARS-CoV-2. Understanding more about the replication of these viruses is imperative in order to effectively find ways to control them.
Collapse
|
13
|
Blahove MR, Carter JR. Flavivirus Persistence in Wildlife Populations. Viruses 2021; 13:v13102099. [PMID: 34696529 PMCID: PMC8541186 DOI: 10.3390/v13102099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022] Open
Abstract
A substantial number of humans are at risk for infection by vector-borne flaviviruses, resulting in considerable morbidity and mortality worldwide. These viruses also infect wildlife at a considerable rate, persistently cycling between ticks/mosquitoes and small mammals and reptiles and non-human primates and humans. Substantially increasing evidence of viral persistence in wildlife continues to be reported. In addition to in humans, viral persistence has been shown to establish in mammalian, reptile, arachnid, and mosquito systems, as well as insect cell lines. Although a considerable amount of research has centered on the potential roles of defective virus particles, autophagy and/or apoptosis-induced evasion of the immune response, and the precise mechanism of these features in flavivirus persistence have yet to be elucidated. In this review, we present findings that aid in understanding how vector-borne flavivirus persistence is established in wildlife. Research studies to be discussed include determining the critical roles universal flavivirus non-structural proteins played in flaviviral persistence, the advancement of animal models of viral persistence, and studying host factors that allow vector-borne flavivirus replication without destructive effects on infected cells. These findings underscore the viral–host relationships in wildlife animals and could be used to elucidate the underlying mechanisms responsible for the establishment of viral persistence in these animals.
Collapse
|
14
|
Nguyen-Dinh V, Herker E. Ultrastructural Features of Membranous Replication Organelles Induced by Positive-Stranded RNA Viruses. Cells 2021; 10:cells10092407. [PMID: 34572055 PMCID: PMC8464962 DOI: 10.3390/cells10092407] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/12/2021] [Accepted: 09/02/2021] [Indexed: 11/25/2022] Open
Abstract
All intracellular pathogens critically depend on host cell organelles and metabolites for successful infection and replication. One hallmark of positive-strand RNA viruses is to induce alterations of the (endo)membrane system in order to shield their double-stranded RNA replication intermediates from detection by the host cell’s surveillance systems. This spatial seclusion also allows for accruing host and viral factors and building blocks required for efficient replication of the genome and prevents access of antiviral effectors. Even though the principle is iterated by almost all positive-strand RNA viruses infecting plants and animals, the specific structure and the organellar source of membranes differs. Here, we discuss the characteristic ultrastructural features of the virus-induced membranous replication organelles in plant and animal cells and the scientific progress gained by advanced microscopy methods.
Collapse
|
15
|
Mlera L, Offerdahl DK, Dorward DW, Carmody A, Chiramel AI, Best SM, Bloom ME. The liver X receptor agonist LXR 623 restricts flavivirus replication. Emerg Microbes Infect 2021; 10:1378-1389. [PMID: 34162308 PMCID: PMC8259867 DOI: 10.1080/22221751.2021.1947749] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The vector-borne flaviviruses (VBFVs) are well known for causing great misery and death in humans worldwide. The VBFVs include those transmitted by mosquitos, such as Zika virus (ZIKV), dengue virus; and those transmitted by ticks including the tick-borne flavivirus serocomplex and Powassan virus (POWV). Two of our recent reports showed that intracranial POWV infection in the reservoir host, Peromyscus leucopus, was restricted and caused no overt clinical disease. Several modes of analyses suggested activation of the LXR pathway. Activation of the LXR pathway leads to increased efflux of cholesterol from cells and consequent disturbances in membrane biogenesis. Because VBFV replication is dependent on membrane biogenesis, we evaluated the effect of an LXR agonist (LXR623) on POWV and ZIKV infection and observed that the compound impaired permissive replication of both viruses in a human neuroblastoma SK-N-SH cell line. The LXR agonist resulted in failure of the viruses to induce ER expansion and elaborate vesicle formation, suggesting that the efflux of cholesterol was part of the antiviral mechanism. We also observed that the LXR agonist contributed to the mechanism of virus suppression by increased expression of mRNAs encoding for the antiviral cytokines CXCL10, RANTES and IFN1β. In sharp contrast, a LXR antagonist (GSK2033) had no significant effect on VBFV replication. We conclude that LXR623 impairs flavivirus replication by stimulating cellular antiviral factors.
Collapse
Affiliation(s)
- Luwanika Mlera
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, NIAID/NIH, Hamilton, MT, USA
| | - Danielle K Offerdahl
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, NIAID/NIH, Hamilton, MT, USA
| | - David W Dorward
- Microscopy Unit, Research Technologies Branch, NIAID/NIH, Hamilton, MT, USA
| | - Aaron Carmody
- Research Technologies Branch, NIAID/NIH, Hamilton, MT, USA
| | - Abhilash I Chiramel
- Innate Immunity and Pathogenesis Section, Laboratory of Virology, NIAID/NIH, Hamilton, MT, USA
| | - Sonja M Best
- Innate Immunity and Pathogenesis Section, Laboratory of Virology, NIAID/NIH, Hamilton, MT, USA
| | - Marshall E Bloom
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, NIAID/NIH, Hamilton, MT, USA
| |
Collapse
|
16
|
Tran PTH, Asghar N, Johansson M, Melik W. Roles of the Endogenous Lunapark Protein during Flavivirus Replication. Viruses 2021; 13:v13071198. [PMID: 34206552 PMCID: PMC8310331 DOI: 10.3390/v13071198] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/08/2021] [Accepted: 06/17/2021] [Indexed: 12/19/2022] Open
Abstract
The endoplasmic reticulum (ER) of eukaryotic cells is a dynamic organelle, which undergoes continuous remodeling. At the three-way tubular junctions of the ER, the lunapark (LNP) protein acts as a membrane remodeling factor to stabilize these highly curved membrane junctions. In addition, during flavivirus infection, the ER membrane is invaginated to form vesicles (Ve) for virus replication. Thus, LNP may have roles in the generation or maintenance of the Ve during flavivirus infection. In this study, our aim was to characterize the functions of LNP during flavivirus infection and investigate the underlying mechanisms of these functions. To specifically study virus replication, we generated cell lines expressing replicons of West Nile virus (Kunjin strain) or Langat virus. By using these replicon platforms and electron microscopy, we showed that depletion of LNP resulted in reduced virus replication, which is due to its role in the generation of the Ve. By using biochemical assays and high-resolution microscopy, we found that LNP is recruited to the Ve and the protein interacts with the nonstructural protein (NS) 4B. Therefore, these data shed new light on the interactions between flavivirus and host factors during viral replication.
Collapse
|
17
|
Morita E, Suzuki Y. Membrane-Associated Flavivirus Replication Complex-Its Organization and Regulation. Viruses 2021; 13:v13061060. [PMID: 34205058 PMCID: PMC8228428 DOI: 10.3390/v13061060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/02/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Flavivirus consists of a large number of arthropod-borne viruses, many of which cause life-threatening diseases in humans. A characteristic feature of flavivirus infection is to induce the rearrangement of intracellular membrane structure in the cytoplasm. This unique membranous structure called replication organelle is considered as a microenvironment that provides factors required for the activity of the flaviviral replication complex. The replication organelle serves as a place to coordinate viral RNA amplification, protein translation, and virion assembly and also to protect the viral replication complex from the cellular immune defense system. In this review, we summarize the current understanding of how the formation and function of membrane-associated flaviviral replication organelle are regulated by cellular factors.
Collapse
Affiliation(s)
- Eiji Morita
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, 3 Bunkyo-cho, Hirosaki-shi 036-8561, Japan
- Correspondence: (E.M.); (Y.S.); Tel.: +81-172-39-3586 (E.M.); +81-72-684-7367 (Y.S.)
| | - Youichi Suzuki
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki 569-8686, Japan
- Correspondence: (E.M.); (Y.S.); Tel.: +81-172-39-3586 (E.M.); +81-72-684-7367 (Y.S.)
| |
Collapse
|
18
|
Bruno SR, Anathy V. Lung epithelial endoplasmic reticulum and mitochondrial 3D ultrastructure: a new frontier in lung diseases. Histochem Cell Biol 2021; 155:291-300. [PMID: 33598824 PMCID: PMC7889473 DOI: 10.1007/s00418-020-01950-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2020] [Indexed: 12/15/2022]
Abstract
It has long been appreciated that the endoplasmic reticulum (ER) and mitochondria, organelles important for regular cell function and survival, also play key roles in pathogenesis of various lung diseases, including asthma, fibrosis, and infections. Alterations in processes regulated within these organelles, including but not limited to protein folding in the ER and oxidative phosphorylation in the mitochondria, are important in disease pathogenesis. In recent years it has also become increasingly apparent that organelle structure dictates function. It is now clear that organelles must maintain precise organization and localization for proper function. Newer microscopy capabilities have allowed the scientific community to reveal, via 3D imaging, that the structure of these organelles and their interactions with each other are a main component of regulating function and, therefore, effects on the disease state. In this review, we will examine how 3D imaging through techniques could allow advancements in knowledge of how the ER and mitochondria function and the roles they may play in lung epithelia in progression of lung disease.
Collapse
Affiliation(s)
- Sierra R Bruno
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, 149 Beaumont Ave, Burlington, VT, 05405, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, 149 Beaumont Ave, Burlington, VT, 05405, USA.
| |
Collapse
|
19
|
Multiscale Electron Microscopy for the Study of Viral Replication Organelles. Viruses 2021; 13:v13020197. [PMID: 33525547 PMCID: PMC7912242 DOI: 10.3390/v13020197] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 11/17/2022] Open
Abstract
During infection with positive-strand RNA viruses, viral RNA synthesis associates with modified intracellular membranes that form unique and captivating structures in the cytoplasm of the infected cell. These viral replication organelles (ROs) play a key role in the replicative cycle of important human pathogens like coronaviruses, enteroviruses, or flaviviruses. From their discovery to date, progress in our understanding of viral ROs has closely followed new developments in electron microscopy (EM). This review gives a chronological account of this progress and an introduction to the different EM techniques that enabled it. With an ample repertoire of imaging modalities, EM is nowadays a versatile technique that provides structural and functional information at a wide range of scales. Together with well-established approaches like electron tomography or labeling methods, we examine more recent developments, such as volume scanning electron microscopy (SEM) and in situ cryotomography, which are only beginning to be applied to the study of viral ROs. We also highlight the first cryotomography analyses of viral ROs, which have led to the discovery of macromolecular complexes that may serve as RO channels that control the export of newly-made viral RNA. These studies are key first steps towards elucidating the macromolecular complexity of viral ROs.
Collapse
|
20
|
Kendall BL, Grabowski JM, Rosenke R, Pulliam M, Long DR, Scott DP, Offerdahl DK, Bloom ME. Characterization of flavivirus infection in salivary gland cultures from male Ixodes scapularis ticks. PLoS Negl Trop Dis 2020; 14:e0008683. [PMID: 33017410 PMCID: PMC7561187 DOI: 10.1371/journal.pntd.0008683] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 10/15/2020] [Accepted: 08/05/2020] [Indexed: 01/01/2023] Open
Abstract
Infected Ixodes scapularis (black-legged tick) transmit a host of serious pathogens via their bites, including Borrelia burgdorferi, Babesia microti, and tick-borne flaviviruses (TBFVs), such as Powassan virus (POWV). Although the role of female I. scapularis ticks in disease transmission is well characterized, the role of male ticks is poorly understood. Because the pathogens are delivered in tick saliva, we studied the capacity of male salivary glands (SGs) to support virus replication. Ex vivo cultures of SGs from unfed male I. scapularis were viable for more than a week and maintained the characteristic tissue architecture of lobular ducts and acini. When SG cultures were infected with the TBFVs Langat virus (LGTV) or POWV lineage II (deer tick virus), the production of infectious virus was demonstrated. Using a green fluorescent protein-tagged LGTV and confocal microscopy, we demonstrated LGTV infection within SG acinus types II and III. The presence of LGTV in the acini and lobular ducts of the cultures was also shown via immunohistochemistry. Furthermore, the identification by in situ hybridization of both positive and negative strand LGTV RNA confirmed that the virus was indeed replicating. Finally, transmission electron microscopy of infected SGs revealed virus particles packaged in vesicles or vacuoles adjacent to acinar lumina. These studies support the concept that SGs of male I. scapularis ticks support replication of TBFVs and may play a role in virus transmission, and further refine a useful model system for developing countermeasures against this important group of pathogens. Powassan disease has greatly increased in frequency since its discovery in Powassan, Ontario in 1958. Powassan virus (lineage I; POWV) and Powassan virus lineage II (deer tick virus; DTV) are endemic to North America and there were 133 reported cases between 2009 and 2018, the majority since 2016. Nymphal and adult Ixodes scapularis ticks are thought to be the primary vectors of POWV/DTV to humans. However, little is known regarding DTV infection of male Ixodes ticks or their potential as vectors. In this study we characterized LGTV, a model tick-borne flavivirus, and DTV infection and propagation in male I. scapularis salivary gland cultures using an ex vivo organ culture system. This work provides insight into potential flavivirus transmission by the male I. scapularis tick.
Collapse
Affiliation(s)
- Benjamin L. Kendall
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID/NIH, Hamilton, MT, United States of America
| | - Jeffrey M. Grabowski
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID/NIH, Hamilton, MT, United States of America
- * E-mail: , (JMG); (MEB)
| | - Rebecca Rosenke
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, NIAID/NIH, Hamilton, MT, United States of America
| | - Mikayla Pulliam
- Microscopy Unit, Research and Technologies Branch, Rocky Mountain Laboratories, NIAID/NIH, Hamilton, MT, United States of America
| | - Daniel R. Long
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, NIAID/NIH, Hamilton, MT, United States of America
| | - Dana P. Scott
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, NIAID/NIH, Hamilton, MT, United States of America
| | - Danielle K. Offerdahl
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID/NIH, Hamilton, MT, United States of America
| | - Marshall E. Bloom
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID/NIH, Hamilton, MT, United States of America
- * E-mail: , (JMG); (MEB)
| |
Collapse
|
21
|
Kirsch JM, Mlera L, Offerdahl DK, VanSickle M, Bloom ME. Tick-Borne Flaviviruses Depress AKT Activity during Acute Infection by Modulating AKT1/2. Viruses 2020; 12:v12101059. [PMID: 32977414 PMCID: PMC7598186 DOI: 10.3390/v12101059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/22/2022] Open
Abstract
Tick-borne flaviviruses (TBFVs) are reemerging public health threats. To develop therapeutics against these pathogens, increased understanding of their interactions with the mammalian host is required. The PI3K-AKT pathway has been implicated in TBFV persistence, but its role during acute virus infection remains poorly understood. Previously, we showed that Langat virus (LGTV)-infected HEK 293T cells undergo a lytic crisis with a few surviving cells that become persistently infected. We also observed that AKT2 mRNA is upregulated in cells persistently infected with TBFV. Here, we investigated the virus-induced effects on AKT expression over the course of acute LGTV infection and found that total phosphorylated AKT (pAKT), AKT1, and AKT2 decrease over time, but AKT3 increases dramatically. Furthermore, cells lacking AKT1 or AKT2 were more resistant to LGTV-induced cell death than wild-type cells because they expressed higher levels of pAKT and antiapoptotic proteins, such as XIAP and survivin. The differential modulation of AKT by LGTV may be a mechanism by which viral persistence is initiated, and our results demonstrate a complicated manipulation of host pathways by TBFVs.
Collapse
|
22
|
Kaufman F, Dostálková A, Pekárek L, Thanh TD, Kapisheva M, Hadravová R, Bednárová L, Novotný R, Křížová I, Černý J, Grubhoffer L, Ruml T, Hrabal R, Rumlová M. Characterization and in vitro assembly of tick-borne encephalitis virus C protein. FEBS Lett 2020; 594:1989-2004. [PMID: 32510601 DOI: 10.1002/1873-3468.13857] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/12/2020] [Accepted: 05/17/2020] [Indexed: 01/10/2023]
Abstract
Tick-borne encephalitis virus (TBEV), a member of flaviviruses, represents a serious health threat by causing human encephalitis mainly in central and eastern Europe, Russia, and northeastern Asia. As no specific therapy is available, there is an urgent need to understand all steps of the TBEV replication cycle at the molecular level. One of the critical events is the packaging of flaviviral genomic RNA by TBEV C protein to form a nucleocapsid. We purified recombinant TBEV C protein and used a combination of physical-chemical approaches, such as size-exclusion chromatography, circular dichroism, NMR spectroscopies, and transmission electron microscopy, to analyze its structural stability and its ability to dimerize/oligomerize. We compared the ability of TBEV C protein to assemble in vitro into a nucleocapsid-like structure with that of dengue C protein.
Collapse
Affiliation(s)
- Filip Kaufman
- Department of Biotechnology, University of Chemistry and Technology, Prague, Prague, Czech Republic
| | - Alžběta Dostálková
- Department of Biotechnology, University of Chemistry and Technology, Prague, Prague, Czech Republic
| | - Lukáš Pekárek
- Department of Biotechnology, University of Chemistry and Technology, Prague, Prague, Czech Republic
| | - Tung Dinh Thanh
- Department of Biotechnology, University of Chemistry and Technology, Prague, Prague, Czech Republic
| | - Marina Kapisheva
- Department of Biotechnology, University of Chemistry and Technology, Prague, Prague, Czech Republic
| | - Romana Hadravová
- Department of Biotechnology, University of Chemistry and Technology, Prague, Prague, Czech Republic.,Institute of Organic Chemistry and Biochemistry (IOCB) Research Centre & Gilead Sciences, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Lucie Bednárová
- Institute of Organic Chemistry and Biochemistry (IOCB) Research Centre & Gilead Sciences, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Radim Novotný
- Department of Biotechnology, University of Chemistry and Technology, Prague, Prague, Czech Republic.,NMR Laboratory, University of Chemistry and Technology, Prague, Prague, Czech Republic
| | - Ivana Křížová
- Department of Biotechnology, University of Chemistry and Technology, Prague, Prague, Czech Republic
| | - Jiří Černý
- Faculty of Tropical AgriSciences, Czech University of Life Sciences, Prague, Prague, Czech Republic
| | - Libor Grubhoffer
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague, Czech Republic
| | - Richard Hrabal
- NMR Laboratory, University of Chemistry and Technology, Prague, Prague, Czech Republic
| | - Michaela Rumlová
- Department of Biotechnology, University of Chemistry and Technology, Prague, Prague, Czech Republic
| |
Collapse
|
23
|
Chen C, Han X, Chen C, Wang F, Huang J, Zhang L, Wang Z, Yang H. Crystal structure of the NS3 helicase of tick-borne encephalitis virus. Biochem Biophys Res Commun 2020; 528:601-606. [PMID: 32505343 DOI: 10.1016/j.bbrc.2020.05.138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 01/07/2023]
Abstract
Tick-borne encephalitis virus (TBEV) is a positive-sense single-stranded RNA virus belonging to the genus Flavivirus in Flaviviridae. It can cause the server infectious diseases named tick-borne encephalitis (TBE), which is characterized by paralysis and epilepsy. However, no effective treatment for TBE has been developed targeting TBEV. The NS3 helicase from TBEV plays an essential role in viral replication, which makes it an important target for drug design. In this study, the crystal structure of TBEV NS3 helicase has been determined to the resolution of 2.14 Å. Subsequent alignment with homologous structures reveals that the NTP binding site and RNA-binding sites are located in motifs Ⅱ and Ⅵ of NS3 and the critical residues for binding are conserved across species in the genus, while the distinct conformation transition implies that the TBEV helicase need a different local rearrangement. This study demonstrates the key atomic-level features of TBEV helicase and provides basis for the design of antiviral drugs targeting TBEV helicase.
Collapse
Affiliation(s)
- Chen Chen
- School of Life Sciences, Tianjin University, Tianjin, China.
| | - Xu Han
- School of Life Sciences, Tianjin University, Tianjin, China.
| | - Cheng Chen
- School of Life Sciences, Tianjin University, Tianjin, China; Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China.
| | - Fenghua Wang
- School of Life Sciences, Tianjin University, Tianjin, China.
| | - Jinhai Huang
- School of Life Sciences, Tianjin University, Tianjin, China.
| | - Lei Zhang
- School of Life Sciences, Tianjin University, Tianjin, China.
| | - Zefang Wang
- School of Life Sciences, Tianjin University, Tianjin, China; Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China.
| | - Haitao Yang
- School of Life Sciences, Tianjin University, Tianjin, China; Tianjin International Joint Academy of Biotechnology and Medicine, Tianjin, China.
| |
Collapse
|
24
|
Lewy TG, Offerdahl DK, Grabowski JM, Kellman E, Mlera L, Chiramel A, Bloom ME. PERK-Mediated Unfolded Protein Response Signaling Restricts Replication of the Tick-Borne Flavivirus Langat Virus. Viruses 2020; 12:v12030328. [PMID: 32197325 PMCID: PMC7150897 DOI: 10.3390/v12030328] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/05/2020] [Accepted: 03/13/2020] [Indexed: 02/07/2023] Open
Abstract
The unfolded protein response (UPR) maintains protein-folding homeostasis in the endoplasmic reticulum (ER) and has been implicated as both beneficial and detrimental to flavivirus infection. Protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), a sensor of the UPR, is commonly associated with antiviral effects during mosquito-borne flavivirus (MBFV) infection, but its relation to tick-borne flavivirus (TBFV) infection remains largely unexplored. In this study, we identified changes in UPR and autophagic activity during Langat virus (LGTV) infection. LGTV robustly activated UPR and altered autophagic flux. Knockdown of endogenous PERK in human cells resulted in increased LGTV replication, but not that of closely related Powassan virus (POWV). Finally, on examining changes in protein levels of components associated with UPR and autophagy in the absence of PERK, we could show that LGTV-infected cells induced UPR but did not lead to expression of C/EBP homologous protein (CHOP), an important downstream transcription factor of multiple stress pathways. From these data, we hypothesize that LGTV can antagonize other kinases that target eukaryotic initiation factor 2α (eIF2α), but not PERK, implicating PERK as a potential mediator of intrinsic immunity. This effect was not apparent for POWV, a more pathogenic TBFV, suggesting it may be better equipped to mitigate the antiviral effects of PERK.
Collapse
Affiliation(s)
- Tyler G. Lewy
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID/NIH, 903 S. 4th St, Hamilton, MT 59840, USA; (T.G.L.); (D.K.O.); (E.K.); (L.M.)
| | - Danielle K. Offerdahl
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID/NIH, 903 S. 4th St, Hamilton, MT 59840, USA; (T.G.L.); (D.K.O.); (E.K.); (L.M.)
| | - Jeffrey M. Grabowski
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID/NIH, 903 S. 4th St, Hamilton, MT 59840, USA; (T.G.L.); (D.K.O.); (E.K.); (L.M.)
- Correspondence: (J.M.G.); (M.E.B.); Tel.: +1-406-375-9789 (J.M.G.); +1-406-375-9707 (M.E.B.)
| | - Eliza Kellman
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID/NIH, 903 S. 4th St, Hamilton, MT 59840, USA; (T.G.L.); (D.K.O.); (E.K.); (L.M.)
| | - Luwanika Mlera
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID/NIH, 903 S. 4th St, Hamilton, MT 59840, USA; (T.G.L.); (D.K.O.); (E.K.); (L.M.)
| | - Abhilash Chiramel
- Innate Immunity and Pathogenesis Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID/NIH, 903 S. 4th St, Hamilton, MT 59840, USA;
| | - Marshall E. Bloom
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID/NIH, 903 S. 4th St, Hamilton, MT 59840, USA; (T.G.L.); (D.K.O.); (E.K.); (L.M.)
- Correspondence: (J.M.G.); (M.E.B.); Tel.: +1-406-375-9789 (J.M.G.); +1-406-375-9707 (M.E.B.)
| |
Collapse
|
25
|
Bagchi P. Endoplasmic reticulum in viral infection. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 350:265-284. [PMID: 32138901 DOI: 10.1016/bs.ircmb.2019.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Virus exploits host cellular machinery to replicate and form new viral progeny and endoplasmic reticulum (ER) plays central role in the interplay between virus and host cell. Here I will discuss how cellular functions of ER being utilized by viruses from different families during different stages of pathogenesis. Flow of knowledge related to this area of research based on interdisciplinary approach, using biochemical and cell biological assays coupled with advanced microscopy strategies, is pushing our understanding of the virus-ER interaction during infection to the next level.
Collapse
Affiliation(s)
- Parikshit Bagchi
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
26
|
The Porcine Deltacoronavirus Replication Organelle Comprises Double-Membrane Vesicles and Zippered Endoplasmic Reticulum with Double-Membrane Spherules. Viruses 2019; 11:v11111030. [PMID: 31694296 PMCID: PMC6893519 DOI: 10.3390/v11111030] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 01/09/2023] Open
Abstract
Porcine deltacoronavirus (PDCoV) was first identified in Hong Kong in 2012 from samples taken from pigs in 2009. PDCoV was subsequently identified in the USA in 2014 in pigs with a history of severe diarrhea. The virus has now been detected in pigs in several countries around the world. Following the development of tissue culture adapted strains of PDCoV, it is now possible to address questions regarding virus-host cell interactions for this genera of coronavirus. Here, we presented a detailed study of PDCoV-induced replication organelles. All positive-strand RNA viruses induce the rearrangement of cellular membranes during virus replication to support viral RNA synthesis, forming the replication organelle. Replication organelles for the Alpha-, Beta-, and Gammacoronavirus genera have been characterized. All coronavirus genera induced the formation of double-membrane vesicles (DMVs). In addition, Alpha- and Betacoronaviruses induce the formation of convoluted membranes, while Gammacoronaviruses induce the formation of zippered endoplasmic reticulum (ER) with tethered double-membrane spherules. However, the structures induced by Deltacoronaviruses, particularly the presence of convoluted membranes or double-membrane spherules, are unknown. Initially, the dynamics of PDCoV strain OH-FD22 replication were assessed with the onset of viral RNA synthesis, protein synthesis, and progeny particle release determined. Subsequently, virus-induced membrane rearrangements were identified in infected cells by electron microscopy. As has been observed for all other coronaviruses studied to date, PDCoV replication was found to induce the formation of double-membrane vesicles. Significantly, however, PDCoV replication was also found to induce the formation of regions of zippered endoplasmic reticulum, small associated tethered vesicles, and double-membrane spherules. These structures strongly resemble the replication organelle induced by avian Gammacoronavirus infectious bronchitis virus.
Collapse
|
27
|
Rothan HA, Kumar M. Role of Endoplasmic Reticulum-Associated Proteins in Flavivirus Replication and Assembly Complexes. Pathogens 2019; 8:E148. [PMID: 31547236 PMCID: PMC6789530 DOI: 10.3390/pathogens8030148] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 01/29/2023] Open
Abstract
Flavivirus replication in host cells requires the formation of replication and assembly complexes on the cytoplasmic side of the endoplasmic reticulum (ER) membrane. These complexes consist of an ER membrane, viral proteins, and host proteins. Genome-wide investigations have identified a number of ER multiprotein complexes as vital factors for flavivirus replication. The detailed mechanisms of the role of ER complexes in flavivirus replication are still largely elusive. This review highlights the fact that the ER multiprotein complexes are crucial for the formation of flavivirus replication and assembly complexes, and the ER complexes could be considered as a target for developing successful broad-spectrum anti-flavivirus drugs.
Collapse
Affiliation(s)
- Hussin A Rothan
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA.
| | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
28
|
Sempere RN, Arias A. Establishment of a Cell Culture Model of Persistent Flaviviral Infection: Usutu Virus Shows Sustained Replication during Passages and Resistance to Extinction by Antiviral Nucleosides. Viruses 2019; 11:E560. [PMID: 31212939 PMCID: PMC6630443 DOI: 10.3390/v11060560] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/02/2019] [Accepted: 06/15/2019] [Indexed: 12/30/2022] Open
Abstract
Chronic viral disease constitutes a major global health problem, with several hundred million people affected and an associated elevated number of deaths. An increasing number of disorders caused by human flaviviruses are related to their capacity to establish a persistent infection. Here we show that Usutu virus (USUV), an emerging zoonotic flavivirus linked to sporadic neurologic disease in humans, can establish a persistent infection in cell culture. Two independent lineages of Vero cells surviving USUV lytic infection were cultured over 82 days (41 cell transfers) without any apparent cytopathology crisis associated. We found elevated titers in the supernatant of these cells, with modest fluctuations during passages but no overall tendency towards increased or decreased infectivity. In addition to full-length genomes, viral RNA isolated from these cells at passage 40 revealed the presence of defective genomes, containing different deletions at the 5' end. These truncated transcripts were all predicted to encode shorter polyprotein products lacking membrane and envelope structural proteins, and most of non-structural protein 1. Treatment with different broad-range antiviral nucleosides revealed that USUV is sensitive to these compounds in the context of a persistent infection, in agreement with previous observations during lytic infections. The exposure of infected cells to prolonged treatment (10 days) with favipiravir and/or ribavirin resulted in the complete clearance of infectivity in the cellular supernatants (decrease of ~5 log10 in virus titers and RNA levels), although modest changes in intracellular viral RNA levels were recorded (<2 log10 decrease). Drug withdrawal after treatment day 10 resulted in a relapse in virus titers. These results encourage the use of persistently-infected cultures as a surrogate system in the identification of improved antivirals against flaviviral chronic disease.
Collapse
Affiliation(s)
- Raquel Navarro Sempere
- Life Science & Bioengineering Building, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
- Abiopep Sociedad Limitada, Parque Científico de Murcia, 30100 Murcia, Spain.
| | - Armando Arias
- Life Science & Bioengineering Building, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| |
Collapse
|
29
|
Potokar M, Jorgačevski J, Zorec R. Astrocytes in Flavivirus Infections. Int J Mol Sci 2019; 20:ijms20030691. [PMID: 30736273 PMCID: PMC6386967 DOI: 10.3390/ijms20030691] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/27/2019] [Accepted: 01/29/2019] [Indexed: 12/14/2022] Open
Abstract
Virus infections of the central nervous system (CNS) can manifest in various forms of inflammation, including that of the brain (encephalitis) and spinal cord (myelitis), all of which may have long-lasting deleterious consequences. Although the knowledge of how different viruses affect neural cells is increasing, understanding of the mechanisms by which cells respond to neurotropic viruses remains fragmented. Several virus types have the ability to infect neural tissue, and astrocytes, an abundant and heterogeneous neuroglial cell type and a key element providing CNS homeostasis, are one of the first CNS cell types to get infected. Astrocytes are morphologically closely aligned with neuronal synapses, blood vessels, and ventricle cavities, and thereby have the capacity to functionally interact with neurons and endothelial cells. In this review, we focus on the responses of astrocytes to infection by neurotropic flaviviruses, including tick-borne encephalitis virus (TBEV), Zika virus (ZIKV), West Nile virus (WNV), and Japanese encephalitis virus (JEV), which have all been confirmed to infect astrocytes and cause multiple CNS defects. Understanding these mechanisms may help design new strategies to better contain and mitigate virus- and astrocyte-dependent neuroinflammation.
Collapse
Affiliation(s)
- Maja Potokar
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia.
- Celica BIOMEDICAL, Tehnološki park 24, 1000 Ljubljana, Slovenia.
| | - Jernej Jorgačevski
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia.
- Celica BIOMEDICAL, Tehnološki park 24, 1000 Ljubljana, Slovenia.
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia.
- Celica BIOMEDICAL, Tehnološki park 24, 1000 Ljubljana, Slovenia.
| |
Collapse
|
30
|
Dissecting Flavivirus Biology in Salivary Gland Cultures from Fed and Unfed Ixodes scapularis (Black-Legged Tick). mBio 2019; 10:mBio.02628-18. [PMID: 30696737 PMCID: PMC6355982 DOI: 10.1128/mbio.02628-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tick-borne flaviviruses (TBFVs) are responsible for more than 15,000 human disease cases each year, and Powassan virus lineage 2 (POWV-L2) deer tick virus has been a reemerging threat in North America over the past 20 years. Rapid transmission of TBFVs in particular emphasizes the importance of preventing tick bites, the difficulty in developing countermeasures to prevent transmission, and the importance of understanding TBFV infection in tick salivary glands (SGs). Tick blood feeding is responsible for phenomenal physiological changes and is associated with changes in TBFV multiplication within the tick and in SGs. Using SG cultures from Ixodes scapularis female ticks, the primary aims of this study were to identify cellular localization of virus-like particles in acini of infected SGs from fed and unfed ticks, localization of TBFV infection in infected SGs from fed ticks, and a tick transcript (with associated metabolic function) involved in POWV-L2 infection in SG cultures. The Ixodes scapularis tick transmits a number of pathogens, including tick-borne flaviviruses (TBFVs). In the United States, confirmed human infections with the Powassan virus (POWV) TBFV have a fatality rate of ∼10% and are increasing in incidence. Tick salivary glands (SGs) serve as an organ barrier to TBFV transmission, and little is known regarding the location of TBFV infection in SGs from fed ticks. Previous studies showed I. scapularis vanin (VNN) involved with TBFV infection of I. scapularis ISE6 embryonic cells, suggesting a potential role for this gene. The overall goal of this study was to use SG cultures to compare data on TBFV biology in SGs from fully engorged, replete (fed) ticks and from unfed ticks. TBFV multiplication was higher in SGs from fed ticks than in those from unfed ticks. Virus-like particles were observed only in granular acini of SGs from unfed ticks. The location of TBFV infection of SGs from fed ticks was observed in cells lining lobular ducts and trachea but not observed in acini. Transcript knockdown of VNN decreased POWV multiplication in infected SG cultures from both fed and unfed ticks. This work was the first to identify localization of TBFV multiplication in SG cultures from a fed tick and a tick transcript important for POWV multiplication in the tick SG, an organ critical for TBFV transmission. This research exemplifies the use of SG cultures in deciphering TBFV biology in the tick and as a translational tool for screening and identifying potential tick genes as potential countermeasure targets.
Collapse
|
31
|
Akkaya B, Oya Y, Akkaya M, Al Souz J, Holstein AH, Kamenyeva O, Kabat J, Matsumura R, Dorward DW, Glass DD, Shevach EM. Regulatory T cells mediate specific suppression by depleting peptide-MHC class II from dendritic cells. Nat Immunol 2019; 20:218-231. [PMID: 30643268 PMCID: PMC6402611 DOI: 10.1038/s41590-018-0280-2] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/08/2018] [Indexed: 01/22/2023]
Abstract
T regulatory cells (Tregs) can activate multiple suppressive mechanisms in vitro upon activation via the T cell receptor resulting in antigen-independent suppression. However, it remains unclear whether similar pathways operate in vivo. Here, we found that antigen-specific Tregs activated by dendritic cells (DCs) pulsed with two antigens suppressed Tnaive specific for both cognate and non-cognate antigens in vitro, but only suppressed Tnaive specific for cognate antigen in vivo. Antigen-specific Tregs formed strong interactions with DC resulting in selective inhibition of the binding of Tnaive to cognate antigen, yet allowing bystander Tnaive access. Strong binding resulted in removal of the cognate peptide-MHCII (pMHCII) from the DC surface reducing the capacity of the DC to present antigen. The enhanced binding of Tregs to DC coupled with their capacity to deplete pMHCII represents a novel pathway for Treg-mediated suppression and may be a mechanism by which Tregs maintain immune homeostasis.
Collapse
Affiliation(s)
- Billur Akkaya
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Yoshihiro Oya
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Department of Rheumatology, Allergy & Clinical Immunology, National Hospital Organization Chiba-East National Hospital, Chiba, Japan
| | - Munir Akkaya
- Laboratory of Immunogenetics National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jafar Al Souz
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amanda H Holstein
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Olena Kamenyeva
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Juraj Kabat
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ryutaro Matsumura
- Department of Rheumatology, Allergy & Clinical Immunology, National Hospital Organization Chiba-East National Hospital, Chiba, Japan
| | - David W Dorward
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Labs, Hamilton, MT, USA
| | - Deborah D Glass
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Rapa Therapeutics, Rockville, MD, USA
| | - Ethan M Shevach
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
32
|
Mazeaud C, Freppel W, Chatel-Chaix L. The Multiples Fates of the Flavivirus RNA Genome During Pathogenesis. Front Genet 2018. [PMID: 30564270 DOI: 10.3389/fgene.2018.00595/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
The Flavivirus genus comprises many viruses (including dengue, Zika, West Nile and yellow fever viruses) which constitute important public health concerns worldwide. For several of these pathogens, neither antivirals nor vaccines are currently available. In addition to this unmet medical need, flaviviruses are of particular interest since they constitute an excellent model for the study of spatiotemporal regulation of RNA metabolism. Indeed, with no DNA intermediate or nuclear step, the flaviviral life cycle entirely relies on the cytoplasmic fate of a single RNA species, namely the genomic viral RNA (vRNA) which contains all the genetic information necessary for optimal viral replication. From a single open reading frame, the vRNA encodes a polyprotein which is processed to generate the mature viral proteins. In addition to coding for the viral polyprotein, the vRNA serves as a template for RNA synthesis and is also selectively packaged into newly assembled viral particles. Notably, vRNA translation, replication and encapsidation must be tightly coordinated in time and space via a fine-tuned equilibrium as these processes cannot occur simultaneously and hence, are mutually exclusive. As such, these dynamic processes involve several vRNA secondary and tertiary structures as well as RNA modifications. Finally, the vRNA can be detected as a foreign molecule by cytosolic sensors which trigger upon activation antiviral signaling pathways and the production of antiviral factors such as interferons and interferon-stimulated genes. However, to create an environment favorable to infection, flaviviruses have evolved mechanisms to dampen these antiviral processes, notably through the production of a specific vRNA degradation product termed subgenomic flavivirus RNA (sfRNA). In this review, we discuss the current understanding of the fates of flavivirus vRNA and how this is regulated at the molecular level to achieve an optimal replication within infected cells.
Collapse
Affiliation(s)
- Clément Mazeaud
- Institut National de la Recherche Scientifique, Centre INRS-Institut Armand-Frappier, Laval, QC, Canada
| | - Wesley Freppel
- Institut National de la Recherche Scientifique, Centre INRS-Institut Armand-Frappier, Laval, QC, Canada
| | - Laurent Chatel-Chaix
- Institut National de la Recherche Scientifique, Centre INRS-Institut Armand-Frappier, Laval, QC, Canada
| |
Collapse
|
33
|
Mazeaud C, Freppel W, Chatel-Chaix L. The Multiples Fates of the Flavivirus RNA Genome During Pathogenesis. Front Genet 2018; 9:595. [PMID: 30564270 PMCID: PMC6288177 DOI: 10.3389/fgene.2018.00595] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/15/2018] [Indexed: 12/11/2022] Open
Abstract
The Flavivirus genus comprises many viruses (including dengue, Zika, West Nile and yellow fever viruses) which constitute important public health concerns worldwide. For several of these pathogens, neither antivirals nor vaccines are currently available. In addition to this unmet medical need, flaviviruses are of particular interest since they constitute an excellent model for the study of spatiotemporal regulation of RNA metabolism. Indeed, with no DNA intermediate or nuclear step, the flaviviral life cycle entirely relies on the cytoplasmic fate of a single RNA species, namely the genomic viral RNA (vRNA) which contains all the genetic information necessary for optimal viral replication. From a single open reading frame, the vRNA encodes a polyprotein which is processed to generate the mature viral proteins. In addition to coding for the viral polyprotein, the vRNA serves as a template for RNA synthesis and is also selectively packaged into newly assembled viral particles. Notably, vRNA translation, replication and encapsidation must be tightly coordinated in time and space via a fine-tuned equilibrium as these processes cannot occur simultaneously and hence, are mutually exclusive. As such, these dynamic processes involve several vRNA secondary and tertiary structures as well as RNA modifications. Finally, the vRNA can be detected as a foreign molecule by cytosolic sensors which trigger upon activation antiviral signaling pathways and the production of antiviral factors such as interferons and interferon-stimulated genes. However, to create an environment favorable to infection, flaviviruses have evolved mechanisms to dampen these antiviral processes, notably through the production of a specific vRNA degradation product termed subgenomic flavivirus RNA (sfRNA). In this review, we discuss the current understanding of the fates of flavivirus vRNA and how this is regulated at the molecular level to achieve an optimal replication within infected cells.
Collapse
Affiliation(s)
- Clément Mazeaud
- Institut National de la Recherche Scientifique, Centre INRS-Institut Armand-Frappier, Laval, QC, Canada
| | - Wesley Freppel
- Institut National de la Recherche Scientifique, Centre INRS-Institut Armand-Frappier, Laval, QC, Canada
| | - Laurent Chatel-Chaix
- Institut National de la Recherche Scientifique, Centre INRS-Institut Armand-Frappier, Laval, QC, Canada
| |
Collapse
|
34
|
Vechtova P, Sterbova J, Sterba J, Vancova M, Rego ROM, Selinger M, Strnad M, Golovchenko M, Rudenko N, Grubhoffer L. A bite so sweet: the glycobiology interface of tick-host-pathogen interactions. Parasit Vectors 2018; 11:594. [PMID: 30428923 PMCID: PMC6236881 DOI: 10.1186/s13071-018-3062-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 08/14/2018] [Indexed: 11/10/2022] Open
Abstract
Vector-borne diseases constitute 17% of all infectious diseases in the world; among the blood-feeding arthropods, ticks transmit the highest number of pathogens. Understanding the interactions between the tick vector, the mammalian host and the pathogens circulating between them is the basis for the successful development of vaccines against ticks or the tick-transmitted pathogens as well as for the development of specific treatments against tick-borne infections. A lot of effort has been put into transcriptomic and proteomic analyses; however, the protein-carbohydrate interactions and the overall glycobiology of ticks and tick-borne pathogens has not been given the importance or priority deserved. Novel (bio)analytical techniques and their availability have immensely increased the possibilities in glycobiology research and thus novel information in the glycobiology of ticks and tick-borne pathogens is being generated at a faster pace each year. This review brings a comprehensive summary of the knowledge on both the glycosylated proteins and the glycan-binding proteins of the ticks as well as the tick-transmitted pathogens, with emphasis on the interactions allowing the infection of both the ticks and the hosts by various bacteria and tick-borne encephalitis virus.
Collapse
Affiliation(s)
- Pavlina Vechtova
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic. .,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic.
| | - Jarmila Sterbova
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| | - Jan Sterba
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| | - Marie Vancova
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| | - Ryan O M Rego
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| | - Martin Selinger
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| | - Martin Strnad
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| | - Maryna Golovchenko
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic
| | - Nataliia Rudenko
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic
| | - Libor Grubhoffer
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Branišovská 31, CZ-37005, České Budějovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branišovská 1760, CZ-37005, České Budějovice, Czech Republic
| |
Collapse
|
35
|
Akkaya B, Roesler AS, Miozzo P, Theall BP, Al Souz J, Smelkinson MG, Kabat J, Traba J, Sack MN, Brzostowski JA, Pena M, Dorward DW, Pierce SK, Akkaya M. Increased Mitochondrial Biogenesis and Reactive Oxygen Species Production Accompany Prolonged CD4 + T Cell Activation. THE JOURNAL OF IMMUNOLOGY 2018; 201:3294-3306. [PMID: 30373851 DOI: 10.4049/jimmunol.1800753] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/25/2018] [Indexed: 01/13/2023]
Abstract
Activation of CD4+ T cells to proliferate drives cells toward aerobic glycolysis for energy production while using mitochondria primarily for macromolecular synthesis. In addition, the mitochondria of activated T cells increase production of reactive oxygen species, providing an important second messenger for intracellular signaling pathways. To better understand the critical changes in mitochondria that accompany prolonged T cell activation, we carried out an extensive analysis of mitochondrial remodeling using a combination of conventional strategies and a novel high-resolution imaging method. We show that for 4 d following activation, mouse CD4+ T cells sustained their commitment to glycolysis facilitated by increased glucose uptake through increased expression of GLUT transporters. Despite their limited contribution to energy production, mitochondria were active and showed increased reactive oxygen species production. Moreover, prolonged activation of CD4+ T cells led to increases in mitochondrial content and volume, in the number of mitochondria per cell and in mitochondrial biogenesis. Thus, during prolonged activation, CD4+ T cells continue to obtain energy predominantly from glycolysis but also undergo extensive mitochondrial remodeling, resulting in increased mitochondrial activity.
Collapse
Affiliation(s)
- Billur Akkaya
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Alexander S Roesler
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Pietro Miozzo
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Brandon P Theall
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Jafar Al Souz
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Margery G Smelkinson
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Juraj Kabat
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Javier Traba
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Michael N Sack
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Joseph A Brzostowski
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Mirna Pena
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - David W Dorward
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840
| | - Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Munir Akkaya
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852;
| |
Collapse
|
36
|
Akkaya M, Traba J, Roesler AS, Miozzo P, Akkaya B, Theall BP, Sohn H, Pena M, Smelkinson M, Kabat J, Dahlstrom E, Dorward DW, Skinner J, Sack MN, Pierce SK. Second signals rescue B cells from activation-induced mitochondrial dysfunction and death. Nat Immunol 2018; 19:871-884. [PMID: 29988090 PMCID: PMC6202187 DOI: 10.1038/s41590-018-0156-5] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/11/2018] [Indexed: 12/31/2022]
Abstract
B cells are activated by two temporally distinct signals, the first provided by antigen binding to the B cell antigen receptor (BCR) and the second by T helper cells. Here we show that B cells responded to antigen by rapidly increasing metabolic activity including both oxidative phosphorylation and glycolysis. In the absence of a second signal B cells progressively lost mitochondrial function and glycolytic capacity leading to apoptosis. Mitochondrial dysfunction was a result of the gradual accumulation of intracellular calcium through calcium response activated calcium channels that was preventable for approximately nine hours after B cell antigen binding by either T helper cells or Toll-like receptor 9 signaling. Thus, BCR signaling appears to activate a metabolic program that imposes a limited time window in which B cells either receive a second signal and survive or are eliminated.
Collapse
Affiliation(s)
- Munir Akkaya
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| | - Javier Traba
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander S Roesler
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.,Duke University School of Medicine, Durham, NC, USA
| | - Pietro Miozzo
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.,University of Massachusetts Medical School, Worcester, MA, USA
| | - Billur Akkaya
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brandon P Theall
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Haewon Sohn
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Mirna Pena
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Margery Smelkinson
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Juraj Kabat
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Eric Dahlstrom
- Genomics Unit, Rocky Mountain Laboratories, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - David W Dorward
- Microscopy Unit, Rocky Mountain Laboratories, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jeff Skinner
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Michael N Sack
- Laboratory of Mitochondrial Biology and Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
37
|
Pulkkinen LIA, Butcher SJ, Anastasina M. Tick-Borne Encephalitis Virus: A Structural View. Viruses 2018; 10:v10070350. [PMID: 29958443 PMCID: PMC6071267 DOI: 10.3390/v10070350] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 06/25/2018] [Accepted: 06/27/2018] [Indexed: 12/11/2022] Open
Abstract
Tick-borne encephalitis virus (TBEV) is a growing health concern. It causes a severe disease that can lead to permanent neurological complications or death and the incidence of TBEV infections is constantly rising. Our understanding of TBEV’s structure lags behind that of other flaviviruses, but has advanced recently with the publication of a high-resolution structure of the TBEV virion. The gaps in our knowledge include: aspects of receptor binding, replication and virus assembly. Furthermore, TBEV has mostly been studied in mammalian systems, even though the virus’ interaction with its tick hosts is a central part of its life cycle. Elucidating these aspects of TBEV biology are crucial for the development of TBEV antivirals, as well as the improvement of diagnostics. In this review, we summarise the current structural knowledge on TBEV, bringing attention to the current gaps in our understanding, and propose further research that is needed to truly understand the structural-functional relationship of the virus and its hosts.
Collapse
Affiliation(s)
- Lauri I A Pulkkinen
- HiLIFE-Institute of Biotechnology, University of Helsinki, 00790 Helsinki, Finland.
- Faculty of Biological and Environmental Sciences, University of Helsinki, 00790 Helsinki, Finland.
| | - Sarah J Butcher
- HiLIFE-Institute of Biotechnology, University of Helsinki, 00790 Helsinki, Finland.
- Faculty of Biological and Environmental Sciences, University of Helsinki, 00790 Helsinki, Finland.
| | - Maria Anastasina
- HiLIFE-Institute of Biotechnology, University of Helsinki, 00790 Helsinki, Finland.
- Faculty of Biological and Environmental Sciences, University of Helsinki, 00790 Helsinki, Finland.
| |
Collapse
|
38
|
Lindqvist R, Upadhyay A, Överby AK. Tick-Borne Flaviviruses and the Type I Interferon Response. Viruses 2018; 10:E340. [PMID: 29933625 PMCID: PMC6071234 DOI: 10.3390/v10070340] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/15/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022] Open
Abstract
Flaviviruses are globally distributed pathogens causing millions of human infections every year. Flaviviruses are arthropod-borne viruses and are mainly transmitted by either ticks or mosquitoes. Mosquito-borne flaviviruses and their interactions with the innate immune response have been well-studied and reviewed extensively, thus this review will discuss tick-borne flaviviruses and their interactions with the host innate immune response.
Collapse
Affiliation(s)
- Richard Lindqvist
- Department of Clinical Microbiology, Virology, Umeå University, SE-90185 Umeå, Sweden.
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, SE-90187 Umeå, Sweden.
- Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187 Umeå, Sweden.
| | - Arunkumar Upadhyay
- Department of Clinical Microbiology, Virology, Umeå University, SE-90185 Umeå, Sweden.
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, SE-90187 Umeå, Sweden.
- Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187 Umeå, Sweden.
| | - Anna K Överby
- Department of Clinical Microbiology, Virology, Umeå University, SE-90185 Umeå, Sweden.
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, SE-90187 Umeå, Sweden.
- Umeå Centre for Microbial Research (UCMR), Umeå University, SE-90187 Umeå, Sweden.
| |
Collapse
|
39
|
Liebscher S, Ambrose RL, Aktepe TE, Mikulasova A, Prier JE, Gillespie LK, Lopez-Denman AJ, Rupasinghe TWT, Tull D, McConville MJ, Mackenzie JM. Phospholipase A2 activity during the replication cycle of the flavivirus West Nile virus. PLoS Pathog 2018; 14:e1007029. [PMID: 29709018 PMCID: PMC5945048 DOI: 10.1371/journal.ppat.1007029] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 05/10/2018] [Accepted: 04/15/2018] [Indexed: 02/06/2023] Open
Abstract
Positive-sense RNA virus intracellular replication is intimately associated with membrane platforms that are derived from host organelles and comprised of distinct lipid composition. For flaviviruses, such as West Nile virus strain Kunjin virus (WNVKUN) we have observed that these membrane platforms are derived from the endoplasmic reticulum and are rich in (at least) cholesterol. To extend these studies and identify the cellular lipids critical for WNVKUN replication we utilized a whole cell lipidomics approach and revealed an elevation in phospholipase A2 (PLA2) activity to produce lyso-phosphatidylcholine (lyso-PChol). We observed that the PLA2 enzyme family is activated in WNVKUN-infected cells and the generated lyso-PChol lipid moieties are sequestered to the subcellular sites of viral replication. The requirement for lyso-PChol was confirmed using chemical inhibition of PLA2, where WNVKUN replication and production of infectious virus was duly affected in the presence of the inhibitors. Importantly, we could rescue chemical-induced inhibition with the exogenous addition of lyso-PChol species. Additionally, electron microscopy results indicate that lyso-PChol appears to contribute to the formation of the WNVKUN membranous replication complex (RC); particularly affecting the morphology and membrane curvature of vesicles comprising the RC. These results extend our current understanding of how flaviviruses manipulate lipid homeostasis to favour their own intracellular replication. Positive-sense RNA viruses remodel the host cytoplasmic membrane architecture to induce the formation of membranous organelles termed viral replication complexes. These complexes aid the virus in providing a more efficient microenvironment for replication but additionally shield immune-stimulatory molecules from the immune response. In this report we have performed whole cell lipidomic approaches to identify a key role for the host phospholipase A2 enzyme family in generating lyso-phospholipids to remodel cellular membranes and shape the West Nile virus (WNV) replication complex. We observed elevated PLA2 activity levels in WNV-infected cell cultures from mammalian as well as arthropod origins suggesting a generic requirement of phospholipid hydrolysis for flavivirus replication. Furthermore, we found that chemical inhibition of these enzymes severely affected the ability of WNV to replicate in cells, and we could attribute this defect to an altered ultrastructural morphology of the viral replication complex. This study provides evidence for a mechanism for the biogenesis of the flavivirus replication complex and the specific utilisation of a host lipid to invoke specific membrane curvature, generating a crucial membrane organelle required for efficient virus replication.
Collapse
Affiliation(s)
- Susann Liebscher
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Rebecca L. Ambrose
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Turgut E. Aktepe
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Andrea Mikulasova
- Department of Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Julia E. Prier
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Leah K. Gillespie
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Adam J. Lopez-Denman
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Thusitha W. T. Rupasinghe
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, Department of Biochemistry and Molecular Biology at University of Melbourne, Melbourne, VIC, Australia
| | - Dedreia Tull
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, Department of Biochemistry and Molecular Biology at University of Melbourne, Melbourne, VIC, Australia
| | - Malcolm J. McConville
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, Department of Biochemistry and Molecular Biology at University of Melbourne, Melbourne, VIC, Australia
| | - Jason M. Mackenzie
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- * E-mail:
| |
Collapse
|
40
|
Liu J, Kline BA, Kenny TA, Smith DR, Soloveva V, Beitzel B, Pang S, Lockett S, Hess HF, Palacios G, Kuhn JH, Sun MG, Zeng X. A novel sheet-like virus particle array is a hallmark of Zika virus infection. Emerg Microbes Infect 2018; 7:69. [PMID: 29691373 PMCID: PMC5915449 DOI: 10.1038/s41426-018-0071-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 02/08/2018] [Accepted: 03/25/2018] [Indexed: 11/08/2022]
Abstract
Zika virus (ZIKV) is an emerging flavivirus that caused thousands of human infections in recent years. Compared to other human flaviviruses, ZIKV replication is not well understood. Using fluorescent, transmission electron, and focused ion beam-scanning electron microscopy, we examined ZIKV replication dynamics in Vero 76 cells and in the brains of infected laboratory mice. We observed the progressive development of a perinuclear flaviviral replication factory both in vitro and in vivo. In vitro, we illustrated the ZIKV lifecycle from particle cell entry to egress. ZIKV particles assembled and aggregated in an induced convoluted membrane structure and ZIKV strain-specific membranous vesicles. While most mature virus particles egressed via membrane budding, some particles also likely trafficked through late endosomes and egressed through membrane abscission. Interestingly, we consistently observed a novel sheet-like virus particle array consisting of a single layer of ZIKV particles. Our study further defines ZIKV replication and identifies a novel hallmark of ZIKV infection.
Collapse
Affiliation(s)
- Jun Liu
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, 21702, USA
| | - Brandon A Kline
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, 21702, USA
| | - Tara A Kenny
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, 21702, USA
| | - Darci R Smith
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, 21702, USA
| | - Veronica Soloveva
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, 21702, USA
| | - Brett Beitzel
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, 21702, USA
| | - Song Pang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, 20147, USA
| | - Stephen Lockett
- Optical Microscopy and Analysis Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Harald F Hess
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, 20147, USA
| | - Gustavo Palacios
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, 21702, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, 21702, USA
| | - Mei G Sun
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, 21702, USA.
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, 21702, USA.
| |
Collapse
|
41
|
Akkaya M, Akkaya B, Kim AS, Miozzo P, Sohn H, Pena M, Roesler AS, Theall BP, Henke T, Kabat J, Lu J, Dorward DW, Dahlstrom E, Skinner J, Miller LH, Pierce SK. Toll-like receptor 9 antagonizes antibody affinity maturation. Nat Immunol 2018; 19:255-266. [PMID: 29476183 PMCID: PMC5839995 DOI: 10.1038/s41590-018-0052-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 01/10/2018] [Indexed: 12/21/2022]
Abstract
Key events in T cell-dependent antibody responses, including affinity maturation, are dependent on the B cell’s presentation of antigen to helper T cells at critical check points in germinal center formation in secondary lymphoid organs. Here we show that Toll-like receptor 9 (TLR9) signaling blocked the ability of antigen-specific B cells to capture, process and present antigen and to activate antigen-specific helper T cells in vitro. In a mouse model in vivo and in a human clinical trial the TLR9 agonist, CpG, enhanced the magnitude of the antibody response to a protein vaccine but failed to promote affinity maturation. Thus, TLR9 signaling may enhance antibody titers at the expense of the ability of B cells to engage in germinal center events that are highly dependent on B cells’ antigen capture and presentation.
Collapse
Affiliation(s)
- Munir Akkaya
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Billur Akkaya
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ann S Kim
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Pietro Miozzo
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Haewon Sohn
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Mirna Pena
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Alexander S Roesler
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Brandon P Theall
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Travis Henke
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Juraj Kabat
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jinghua Lu
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - David W Dorward
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Eric Dahlstrom
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jeff Skinner
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Louis H Miller
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
42
|
Stefanik M, Formanova P, Bily T, Vancova M, Eyer L, Palus M, Salat J, Braconi CT, Zanotto PMDA, Gould EA, Ruzek D. Characterisation of Zika virus infection in primary human astrocytes. BMC Neurosci 2018; 19:5. [PMID: 29463209 PMCID: PMC5820785 DOI: 10.1186/s12868-018-0407-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 02/16/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The recent Zika virus (ZIKV) outbreak has linked ZIKV with microcephaly and other central nervous system pathologies in humans. Astrocytes are among the first cells to respond to ZIKV infection in the brain and are also targets for virus infection. In this study, we investigated the interaction between ZIKV and primary human brain cortical astrocytes (HBCA). RESULTS HBCAs were highly sensitive to representatives of both Asian and African ZIKV lineages and produced high viral yields. The infection was associated with limited immune cytokine/chemokine response activation; the highest increase of expression, following infection, was seen in CXCL-10 (IP-10), interleukin-6, 8, 12, and CCL5 (RANTES). Ultrastructural changes in the ZIKV-infected HBCA were characterized by electron tomography (ET). ET reconstructions elucidated high-resolution 3D images of the proliferating and extensively rearranged endoplasmic reticulum (ER) containing viral particles and virus-induced vesicles, tightly juxtaposed to collapsed ER cisternae. CONCLUSIONS The results confirm that human astrocytes are sensitive to ZIKV infection and could be a source of proinflammatory cytokines in the ZIKV-infected brain tissue.
Collapse
Affiliation(s)
- Michal Stefanik
- Department of Virology, Veterinary Research Institute, Hudcova 70, 62100, Brno, Czech Republic
| | - Petra Formanova
- Department of Virology, Veterinary Research Institute, Hudcova 70, 62100, Brno, Czech Republic
| | - Tomas Bily
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, 37005, Ceske Budejovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branisovska 31, 37005, Ceske Budejovice, Czech Republic
| | - Marie Vancova
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, 37005, Ceske Budejovice, Czech Republic.,Faculty of Science, University of South Bohemia, Branisovska 31, 37005, Ceske Budejovice, Czech Republic
| | - Ludek Eyer
- Department of Virology, Veterinary Research Institute, Hudcova 70, 62100, Brno, Czech Republic.,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, 37005, Ceske Budejovice, Czech Republic
| | - Martin Palus
- Department of Virology, Veterinary Research Institute, Hudcova 70, 62100, Brno, Czech Republic.,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, 37005, Ceske Budejovice, Czech Republic
| | - Jiri Salat
- Department of Virology, Veterinary Research Institute, Hudcova 70, 62100, Brno, Czech Republic
| | - Carla Torres Braconi
- Laboratory of Molecular Evolution and Bioinformatics, Department of Microbiology, Institute of Microbiology Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Paolo M de A Zanotto
- Laboratory of Molecular Evolution and Bioinformatics, Department of Microbiology, Institute of Microbiology Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Ernest A Gould
- EHESP French School of Public Health, IRD French Institute of Research for Development, EPV UMR_D 190 Emergence des Pathologies Virales, Aix Marseille Université, Marseille, France
| | - Daniel Ruzek
- Department of Virology, Veterinary Research Institute, Hudcova 70, 62100, Brno, Czech Republic. .,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, 37005, Ceske Budejovice, Czech Republic.
| |
Collapse
|
43
|
Grabowski JM, Hill CA. A Roadmap for Tick-Borne Flavivirus Research in the "Omics" Era. Front Cell Infect Microbiol 2017; 7:519. [PMID: 29312896 PMCID: PMC5744076 DOI: 10.3389/fcimb.2017.00519] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/05/2017] [Indexed: 12/30/2022] Open
Abstract
Tick-borne flaviviruses (TBFs) affect human health globally. Human vaccines provide protection against some TBFs, and antivirals are available, yet TBF-specific control strategies are limited. Advances in genomics offer hope to understand the viral complement transmitted by ticks, and to develop disruptive, data-driven technologies for virus detection, treatment, and control. The genome assemblies of Ixodes scapularis, the North American tick vector of the TBF, Powassan virus, and other tick vectors, are providing insights into tick biology and pathogen transmission and serve as nucleation points for expanded genomic research. Systems biology has yielded insights to the response of tick cells to viral infection at the transcript and protein level, and new protein targets for vaccines to limit virus transmission. Reverse vaccinology approaches have moved candidate tick antigenic epitopes into vaccine development pipelines. Traditional drug and in silico screening have identified candidate antivirals, and target-based approaches have been developed to identify novel acaricides. Yet, additional genomic resources are required to expand TBF research. Priorities include genome assemblies for tick vectors, “omic” studies involving high consequence pathogens and vectors, and emphasizing viral metagenomics, tick-virus metabolomics, and structural genomics of TBF and tick proteins. Also required are resources for forward genetics, including the development of tick strains with quantifiable traits, genetic markers and linkage maps. Here we review the current state of genomic research on ticks and tick-borne viruses with an emphasis on TBFs. We outline an ambitious 10-year roadmap for research in the “omics era,” and explore key milestones needed to accomplish the goal of delivering three new vaccines, antivirals and acaricides for TBF control by 2030.
Collapse
Affiliation(s)
- Jeffrey M Grabowski
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Catherine A Hill
- Department of Entomology, Purdue University, West Lafayette, IN, United States.,Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
44
|
Cortese M, Goellner S, Acosta EG, Neufeldt CJ, Oleksiuk O, Lampe M, Haselmann U, Funaya C, Schieber N, Ronchi P, Schorb M, Pruunsild P, Schwab Y, Chatel-Chaix L, Ruggieri A, Bartenschlager R. Ultrastructural Characterization of Zika Virus Replication Factories. Cell Rep 2017; 18:2113-2123. [PMID: 28249158 PMCID: PMC5340982 DOI: 10.1016/j.celrep.2017.02.014] [Citation(s) in RCA: 244] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 01/31/2017] [Accepted: 02/01/2017] [Indexed: 12/20/2022] Open
Abstract
A global concern has emerged with the pandemic spread of Zika virus (ZIKV) infections that can cause severe neurological symptoms in adults and newborns. ZIKV is a positive-strand RNA virus replicating in virus-induced membranous replication factories (RFs). Here we used various imaging techniques to investigate the ultrastructural details of ZIKV RFs and their relationship with host cell organelles. Analyses of human hepatic cells and neural progenitor cells infected with ZIKV revealed endoplasmic reticulum (ER) membrane invaginations containing pore-like openings toward the cytosol, reminiscent to RFs in Dengue virus-infected cells. Both the MR766 African strain and the H/PF/2013 Asian strain, the latter linked to neurological diseases, induce RFs of similar architecture. Importantly, ZIKV infection causes a drastic reorganization of microtubules and intermediate filaments forming cage-like structures surrounding the viral RF. Consistently, ZIKV replication is suppressed by cytoskeleton-targeting drugs. Thus, ZIKV RFs are tightly linked to rearrangements of the host cell cytoskeleton. ZIKV induces ER membrane invaginations similar to Dengue virus ZIKV induces profound alterations of the cytoskeleton Microtubules and intermediate filaments surround the ZIKV replication factory ZIKV replication is sensitive to cytoskeleton-targeting drugs
Collapse
Affiliation(s)
- Mirko Cortese
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, 69120 Heidelberg, Germany
| | - Sarah Goellner
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, 69120 Heidelberg, Germany
| | - Eliana Gisela Acosta
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, 69120 Heidelberg, Germany
| | - Christopher John Neufeldt
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, 69120 Heidelberg, Germany
| | - Olga Oleksiuk
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, 69120 Heidelberg, Germany
| | - Marko Lampe
- Advanced Light Microscopy Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Uta Haselmann
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, 69120 Heidelberg, Germany
| | - Charlotta Funaya
- Electron Microscopy Core Facility, Heidelberg University, 69120 Heidelberg, Germany
| | - Nicole Schieber
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Paolo Ronchi
- Electron Microscopy Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Martin Schorb
- Electron Microscopy Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Priit Pruunsild
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Yannick Schwab
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany; Electron Microscopy Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Laurent Chatel-Chaix
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, 69120 Heidelberg, Germany; Institut National de la Recherche Scientifique, Institut Armand-Frappier, Québec H7V 1B7, Canada
| | - Alessia Ruggieri
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, 69120 Heidelberg, Germany
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Heidelberg University, 69120 Heidelberg, Germany; German Center for Infection Research (DZIF), Heidelberg University, 69120 Heidelberg, Germany.
| |
Collapse
|
45
|
Peromyscus leucopus mouse brain transcriptome response to Powassan virus infection. J Neurovirol 2017; 24:75-87. [PMID: 29147886 PMCID: PMC5790856 DOI: 10.1007/s13365-017-0596-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/29/2017] [Accepted: 10/25/2017] [Indexed: 12/30/2022]
Abstract
Powassan virus (POWV) is a tick-borne Flavivirus responsible for life-threatening encephalitis in North America and some regions of Russia. The ticks that have been reported to transmit the virus belong to the Ixodes species, and they feed on small-to-medium-sized mammals, such as Peromyscus leucopus mice, skunks, and woodchucks. We previously developed a P. leucopus mouse model of POWV infection, and the model is characterized by a lack of clinical signs of disease following intraperitoneal or intracranial inoculation. However, intracranial inoculation results in mild subclinical encephalitis from 5 days post infection (dpi), but the encephalitis resolves by 28 dpi. We used RNA sequencing to profile the P. leucopus mouse brain transcriptome at different time points after intracranial challenge with POWV. At 24 h post infection, 42 genes were significantly differentially expressed and the number peaked to 232 at 7 dpi before declining to 31 at 28 dpi. Using Ingenuity Pathway Analysis, we determined that the genes that were significantly expressed from 1 to 15 dpi were mainly associated with interferon signaling. As a result, many interferon-stimulated genes (ISGs) were upregulated. Some of the ISGs include an array of TRIMs (genes encoding tripartite motif proteins). These results will be useful for the identification of POWV restriction factors.
Collapse
|
46
|
Investigating Tick-borne Flaviviral-like Particles as a Delivery System for Gene Therapy. Curr Ther Res Clin Exp 2017; 88:8-17. [PMID: 30093925 PMCID: PMC6076373 DOI: 10.1016/j.curtheres.2017.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2017] [Indexed: 12/30/2022] Open
Abstract
Background Research on the biogenesis of tick-borne encephalitis virus (TBEV) would benefit gene therapy. Due to specific arrangements of genes along the TBEV genome, its viral-like particles (VLPs) could be exploited as shuttles to deliver their replicon, which carries therapeutic genes, to immune system cells. Objective To develop a flaviviral vector for gene delivery as a part of gene therapy research that can be expressed in secretable VLP suicidal shuttles and provide abundant unique molecular and structural data supporting this gene therapy concept. Method TBEV structural gene constructs of a Swedish Torö strain were cloned into plasmids driven by the promoters CAG and CMV and then transfected into various cell lines, including COS-1 and BHK-21. Time-course sampling of the cells, culture fluid, cell lysate supernatant, and pellet specimens were performed. Western blotting and electron microscopy analyses of collected specimens were used to investigate molecular and structural processing of TBEV structural proteins. Results Western blotting analysis showed differences between promoters in directing the gene expression of the VLPs constructs. The premature flaviviral polypeptides as well as mature VLPs could be traced. Using electron microscopy, the premature and mature VLP accumulation in cellular compartments—and also endoplasmic reticulum proliferation as a virus factory platform—were observed in addition to secreted VLPs. Conclusions The abundant virologic and cellular findings in this study show the natural processing and safety of inserting flaviviral structural genes into suicidal VLP shuttles. Thus, we propose that these VLPs are a suitable gene delivering system model in gene therapy.
Collapse
|
47
|
Grabowski JM, Tsetsarkin KA, Long D, Scott DP, Rosenke R, Schwan TG, Mlera L, Offerdahl DK, Pletnev AG, Bloom ME. Flavivirus Infection of Ixodes scapularis (Black-Legged Tick) Ex Vivo Organotypic Cultures and Applications for Disease Control. mBio 2017; 8:e01255-17. [PMID: 28830948 PMCID: PMC5565970 DOI: 10.1128/mbio.01255-17] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 07/25/2017] [Indexed: 12/21/2022] Open
Abstract
Ixodes scapularis ticks transmit many infectious agents that cause disease, including tick-borne flaviviruses (TBFVs). TBFV infections cause thousands of human encephalitis cases worldwide annually. In the United States, human TBFV infections with Powassan virus (POWV) are increasing and have a fatality rate of 10 to 30%. Additionally, Langat virus (LGTV) is a TBFV of low neurovirulence and is used as a model TBFV. TBFV replication and dissemination within I. scapularis organs are poorly characterized, and a deeper understanding of virus biology in this vector may inform effective countermeasures to reduce TBFV transmission. Here, we describe short-term, I. scapularis organ culture models of TBFV infection. Ex vivo organs were metabolically active for 9 to 10 days and were permissive to LGTV and POWV replication. Imaging and videography demonstrated replication and spread of green fluorescent protein-expressing LGTV in the organs. Immunohistochemical staining confirmed LGTV envelope and POWV protein synthesis within the infected organs. LGTV- and POWV-infected organs produced infectious LGTV and POWV; thus, the ex vivo cultures were suitable for study of virus replication in individual organs. LGTV- and POWV-infected midgut and salivary glands were subjected to double-stranded RNA (dsRNA) transfection with dsRNA to the LGTV 3' untranslated region (UTR), which reduced infectious LGTV and POWV replication, providing a proof-of-concept use of RNA interference in I. scapularis organ cultures to study the effects on TBFV replication. The results contribute important information on TBFV localization within ex vivo I. scapularis organs and provide a significant translational tool for evaluating recombinant, live vaccine candidates and potential tick transcripts and proteins for possible therapeutic use and vaccine development to reduce TBFV transmission.IMPORTANCE Tick-borne flavivirus (TBFV) infections cause neurological and/or hemorrhagic disease in humans worldwide. There are currently no licensed therapeutics or vaccines against Powassan virus (POWV), the only TBFV known to circulate in North America. Evaluating tick vector targets for antitick vaccines directed at reducing TBFV infection within the arthropod vector is a critical step in identifying efficient approaches to controlling TBFV transmission. This study characterized infection of female Ixodes scapularis tick organ cultures of midgut, salivary glands, and synganglion with the low-neurovirulence Langat virus (LGTV) and the more pathogenic POWV. Cell types of specific organs were susceptible to TBFV infection, and a difference in LGTV and POWV replication was noted in TBFV-infected organs. This tick organ culture model of TBFV infection will be useful for various applications, such as screening of tick endogenous dsRNA corresponding to potential control targets within midgut and salivary glands to confirm restriction of TBFV infection.
Collapse
Affiliation(s)
- Jeffrey M Grabowski
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA
| | - Konstantin A Tsetsarkin
- Neurotropic Flaviviruses Section, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Dan Long
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA
| | - Dana P Scott
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA
| | - Rebecca Rosenke
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA
| | - Tom G Schwan
- Laboratory of Zoonotic Pathogens, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA
| | - Luwanika Mlera
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA
| | - Danielle K Offerdahl
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA
| | - Alexander G Pletnev
- Neurotropic Flaviviruses Section, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Marshall E Bloom
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA
| |
Collapse
|
48
|
Lewy TG, Grabowski JM, Bloom ME. BiP: Master Regulator of the Unfolded Protein Response and Crucial Factor in Flavivirus Biology
. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2017; 90:291-300. [PMID: 28656015 PMCID: PMC5482305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Flaviviruses have an intimate relationship with their host cells, utilizing host proteins during replication. Much of viral genome replication and virion assembly occurs on and within the endoplasmic reticulum (ER). As a cellular protein folding hub, the ER provides an ideal environment for flaviviruses to replicate. Flaviviruses can interact with several ER processes, including the unfolded protein response (UPR), a cellular stress mechanism responsible for managing unfolded protein accumulation and ER stress. The UPR can alter the ER environment in several ways, including increasing ER volume and quantity of available chaperones, both of which can favor viral replication. BiP, a chaperone and master regulator of the UPR, has been demonstrated to play a key role in several flavivirus infections. Here we describe what is known in regard to BiP, its implicated role with flavivirus infection, and what remains to be discovered.
Collapse
Affiliation(s)
| | | | - Marshall E. Bloom
- To whom all correspondence should be addressed: Marshall E. Bloom, 903 S. 4th St, Hamilton, MT 59840, Tel: (406) 375-9707, Fax: (406) 375-9640, .
| |
Collapse
|
49
|
Chu J, Smelkinson MG, Dorward DW, Zarember KA, Gallin JI. Early Intracellular Trafficking of Granulibacter bethesdensis in Human Macrophages. Infect Immun 2017; 85:e00847-16. [PMID: 28320834 PMCID: PMC5442635 DOI: 10.1128/iai.00847-16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 03/12/2017] [Indexed: 01/30/2023] Open
Abstract
Granulibacter bethesdensis is a Gram-negative bacterium that infects patients with chronic granulomatous disease (CGD), a primary immunodeficiency marked by a defect in NOX2, the phagocyte NADPH oxidase. Previous studies have shown that NOX2 is essential for killing of G. bethesdensis by neutrophils and monocytes and that the bacteriostatic activity of monocyte-derived macrophages (MDM) requires NOX2 and gamma interferon (IFN-γ) pretreatment. To determine whether G. bethesdensis evades phagolysosomal killing, a host defense pathway intact in both normal and CGD MDM, or whether it occupies a distinct intracellular niche in CGD MDM, we assessed the trafficking patterns of this organism. We observed colocalization of G. bethesdensis with an early endosome antigen 1 (EEA1)-positive compartment, followed by colocalization with lysosome-associated membrane protein 1 (LAMP1)-positive and LysoTracker-positive late phagosomes; these characteristics were similar in both normal and CGD MDM. Despite localization to acidified late phagosomes, viable G. bethesdensis cells were recovered from viable MDM in numbers greater than in the initial input up to 6 days after infection. G. bethesdensis remains, and in some cases appears to divide, within a membrane-bound compartment for the entire 6-day time course. These findings indicate that this organism resists both oxygen-dependent and oxygen-independent phagolysosomal antimicrobial systems of human macrophages.
Collapse
Affiliation(s)
- Jessica Chu
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Margery G Smelkinson
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - David W Dorward
- Electron Microscopy Unit, Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Kol A Zarember
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - John I Gallin
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
50
|
Rossignol ED, Peters KN, Connor JH, Bullitt E. Zika virus induced cellular remodelling. Cell Microbiol 2017; 19. [PMID: 28318141 DOI: 10.1111/cmi.12740] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/14/2017] [Accepted: 03/16/2017] [Indexed: 01/08/2023]
Abstract
Zika virus (ZIKV) has been associated with morbidities such as Guillain-Barré, infant microcephaly, and ocular disease. The spread of this positive-sense, single-stranded RNA virus and its growing public health threat underscore gaps in our understanding of basic ZIKV virology. To advance knowledge of the virus replication cycle within mammalian cells, we use serial section 3-dimensional electron tomography to demonstrate the widespread remodelling of intracellular membranes upon infection with ZIKV. We report extensive structural rearrangements of the endoplasmic reticulum and reveal stages of the ZIKV viral replication cycle. Structures associated with RNA genome replication and virus assembly are observed integrated within the endoplasmic reticulum, and we show viruses in transit through the Golgi apparatus for viral maturation, and subsequent cellular egress. This study characterises in detail the 3-dimensional ultrastructural organisation of the ZIKV replication cycle stages. Our results show close adherence of the ZIKV replication cycle to the existing flavivirus replication paradigm.
Collapse
Affiliation(s)
- Evan D Rossignol
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, USA
| | - Kristen N Peters
- Department of Microbiology and National Emerging Infectious Disease Laboratory, Boston University School of Medicine, Boston, MA, USA
| | - John H Connor
- Department of Microbiology and National Emerging Infectious Disease Laboratory, Boston University School of Medicine, Boston, MA, USA
| | - Esther Bullitt
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|