113 results on '"Pijlman GP"'
Search Results
2. Chikungunya virus-like particles are more immunogenic in a lethal AG129 mouse model compared to glycoprotein El or E2 subunits
- Author
-
Metz, SW, Martina, Byron, van den Doel, Petra, Geertsema, C (Corinne), Osterhaus, Ab, Vlak, JM, Pijlman, GP, Virology, and Immunology
- Subjects
Laboratorium voor Virologie ,SDG 3 - Good Health and Well-being ,insect cells ,expression ,Laboratory of Virology ,virus diseases ,PE&RC - Abstract
Chikungunya virus (CHIKV) causes acute illness characterized by fever and long-lasting arthritic symptoms. The need for a safe and effective vaccine against CHM/infections is on the rise due to on-going vector spread and increasing severity of clinical complications. Here we report the results of a comparative vaccination-challenge experiment in mice using three different vaccine candidates produced in insect cells by recombinant baculoviruses: (i) secreted (s)E1 and (ii) sE2 CHIKV glycoprotein subunits (2 mu g/immunization), and (iii) CHIKV virus-like particles (VLPs) (1 mu g E2 equivalent/immunization). These experiments show that vaccination with two subsequent administrations of 1 mu g of Matrix M adjuvanted CHIN VLPs completely protected AG129 mice from lethal CHIN challenge. Vaccination with El and E2 subunits provided partial protection, with half of the mice surviving but with significantly lower neutralizing antibody titres as compared to the VLP vaccinated mice. This study provides evidence that even a modest neutralizing antibody response is sufficient to protect mice from CHM/infections. Neutralization was the prominent correlate of protection. In addition, CHIKV VLPs provide a superior immune response and protection against CHIICV-induced disease in mice as compared to individual CHIKV-sEl and -sE2 subunits. (C) 2013 Elsevier Ltd. All rights reserved.
- Published
- 2013
3. Induction and suppression of tick cell antiviral RNAi responses by tick-borne flaviviruses
- Author
-
Schnettler, E, Tykalova, H, Watson, M, Sharma, M, Sterken, MG, Obbard, DJ, Lewis, SH, McFarlane, M, Bell-Sakyi, L, Barry, G, Weisheit, S, Best, SM, Kuhn, RJ, Pijlman, GP, Chase-Topping, ME, Gould, EA, Grubhoffer, L, Fazakerley, JK, Kohl, A, Schnettler, E, Tykalova, H, Watson, M, Sharma, M, Sterken, MG, Obbard, DJ, Lewis, SH, McFarlane, M, Bell-Sakyi, L, Barry, G, Weisheit, S, Best, SM, Kuhn, RJ, Pijlman, GP, Chase-Topping, ME, Gould, EA, Grubhoffer, L, Fazakerley, JK, and Kohl, A
- Abstract
Arboviruses are transmitted by distantly related arthropod vectors such as mosquitoes (class Insecta) and ticks (class Arachnida). RNA interference (RNAi) is the major antiviral mechanism in arthropods against arboviruses. Unlike in mosquitoes, tick antiviral RNAi is not understood, although this information is important to compare arbovirus/host interactions in different classes of arbovirus vectos. Using an Ixodes scapularis-derived cell line, key Argonaute proteins involved in RNAi and the response against tick-borne Langat virus (Flaviviridae) replication were identified and phylogenetic relationships characterized. Analysis of small RNAs in infected cells showed the production of virus-derived small interfering RNAs (viRNAs), which are key molecules of the antiviral RNAi response. Importantly, viRNAs were longer (22 nucleotides) than those from other arbovirus vectors and mapped at highest frequency to the termini of the viral genome, as opposed to mosquito-borne flaviviruses. Moreover, tick-borne flaviviruses expressed subgenomic flavivirus RNAs that interfere with tick RNAi. Our results characterize the antiviral RNAi response in tick cells including phylogenetic analysis of genes encoding antiviral proteins, and viral interference with this pathway. This shows important differences in antiviral RNAi between the two major classes of arbovirus vectors, and our data broadens our understanding of arthropod antiviral RNAi.
- Published
- 2014
4. Recombinant Modified Vaccinia Virus Ankara Expressing Glycoprotein E2 of Chikungunya Virus Protects AG129 Mice against Lethal Challenge
- Author
-
van den Doel, Petra, Volz, A, Roose, Jeroen, Sewbalaksing, Varsha, Pijlman, GP, van Middelkoop, I, Duiverman, V, de Wetering, EV, Sutter, G, Osterhaus, Ab, Martina, Byron, van den Doel, Petra, Volz, A, Roose, Jeroen, Sewbalaksing, Varsha, Pijlman, GP, van Middelkoop, I, Duiverman, V, de Wetering, EV, Sutter, G, Osterhaus, Ab, and Martina, Byron
- Abstract
Chikungunya virus (CHIKV) infection is characterized by rash, acute high fever, chills, headache, nausea, photophobia, vomiting, and severe polyarthralgia. There is evidence that arthralgia can persist for years and result in long-term discomfort. Neurologic disease with fatal outcome has been documented, although at low incidences. The CHIKV RNA genome encodes five structural proteins (C, E1, E2, E3 and 6K). The E1 spike protein drives the fusion process within the cytoplasm, while the E2 protein is believed to interact with cellular receptors and therefore most probably constitutes the target of neutralizing antibodies. We have constructed recombinant Modified Vaccinia Ankara (MVA) expressing E3E2, 6KE1, or the entire CHIKV envelope polyprotein cassette E3E26KE1. MVA is an appropriate platform because of its demonstrated clinical safety and its suitability for expression of various heterologous proteins. After completing the immunization scheme, animals were challenged with CHIV-S27. Immunization of AG129 mice with MVAs expressing E2 or E3E26KE1 elicited neutralizing antibodies in all animals and provided 100% protection against lethal disease. In contrast, 75% of the animals immunized with 6KE1 were protected against lethal infection. In conclusion, MVA expressing the glycoprotein E2 of CHIKV represents as an immunogenic and effective candidate vaccine against CHIKV infections.
- Published
- 2014
5. Functional processing and secretion of Chikungunya virus E1 and E2 glycoproteins in insect cells
- Author
-
Metz, SW, Geertsema, C (Corinne), Martina, Byron, Andrade, P, Heldens, JG, van Oers, MM, Goldbach, RW, Vlak, JM, Pijlman, GP, Metz, SW, Geertsema, C (Corinne), Martina, Byron, Andrade, P, Heldens, JG, van Oers, MM, Goldbach, RW, Vlak, JM, and Pijlman, GP
- Abstract
Background: Chikungunya virus (CHIKV) is a mosquito-borne, arthrogenic Alphavirus that causes large epidemics in Africa, South-East Asia and India. Recently, CHIKV has been transmitted to humans in Southern Europe by invading and now established Asian tiger mosquitoes. To study the processing of envelope proteins E1 and E2 and to develop a CHIKV subunit vaccine, C-terminally his-tagged E1 and E2 envelope glycoproteins were produced at high levels in insect cells with baculovirus vectors using their native signal peptides located in CHIKV 6K and E3, respectively. Results: Expression in the presence of either tunicamycin or furin inhibitor showed that a substantial portion of recombinant intracellular E1 and precursor E3E2 was glycosylated, but that a smaller fraction of E3E2 was processed by furin into mature E3 and E2. Deletion of the C-terminal transmembrane domains of E1 and E2 enabled secretion of furin-cleaved, fully processed E1 and E2 subunits, which could then be efficiently purified from cell culture fluid via metal affinity chromatography. Confocal laser scanning microscopy on living baculovirus-infected Sf21 cells revealed that full-length E1 and E2 translocated to the plasma membrane, suggesting similar posttranslational processing of E1 and E2, as in a natural CHIKV infection. Baculovirus-directed expression of E1 displayed fusogenic activity as concluded from syncytia formation. CHIKV-E2 was able to induce neutralizing antibodies in rabbits. Conclusions: Chikungunya virus glycoproteins could be functionally expressed at high levels in insect cells and are properly glycosylated and cleaved by furin. The ability of purified, secreted CHIKV-E2 to induce neutralizing antibodies in rabbits underscores the potential use of E2 in a subunit vaccine to prevent CHIKV infections.
- Published
- 2011
6. Process intensification of the baculovirus expression vector system using a perfusion process with a low multiplicity of infection at high cell concentrations.
- Author
-
Altenburg JJ, Juarez-Garza BE, van Keimpema J, van Oosten L, Pijlman GP, van Oers MM, Wijffels RH, and Martens DE
- Abstract
The emergence of new viruses and the spread of existing pathogens necessitate efficient vaccine production methods. The baculovirus expression vector system (BEVS) is an efficient and scalable system for subunit and virus-like particle vaccine production and gene therapy vectors. However, current production processes are often limited to low cell concentrations (1-4 × 10
6 cells/mL) in fed-batch mode. To improve the volumetric productivity of the BEVS, a medium exchange strategy was investigated. Screening experiments were performed to test baculovirus (expressing green fluorescent protein; GFP) infection and productivity of insect cell cultures infected at high cell concentration (1-2 × 107 cells/mL), showing that infection at high cell concentrations was possible with medium exchange. Next, duplicate perfusion runs with baculovirus infection were performed using a cell concentration upon infection (CCI) of 1.2 × 107 cells/mL and a multiplicity of infection (MOI) of 0.01, reaching a maximum viable cell concentration of 2.8 × 107 cells/mL and a maximum GFP production of 263 mg/L. The volumetric productivity of these perfusion runs was 4.8 times higher than for reference batch processes with a CCI of 3 × 106 cells/mL and an MOI of 1. These results demonstrate that process intensification can be achieved for the BEVS by implementing perfusion, resulting in a higher volumetric productivity., (© 2025 The Author(s). Biotechnology Progress published by Wiley Periodicals LLC on behalf of American Institute of Chemical Engineers.)- Published
- 2025
- Full Text
- View/download PDF
7. Continuous Production of Influenza VLPs Using IC-BEVS and Multi-Stage Bioreactors.
- Author
-
Correia R, Zotler T, Ferraz F, Fernandes B, Graça M, Pijlman GP, Alves PM, and Roldão A
- Abstract
The insect cell-baculovirus expression vector system (IC-BEVS) has been an asset to produce biologics for over 30 years. With the current trend in biotechnology shifting toward process intensification and integration, developing intensified processes such as continuous production is crucial to hold this platform as a suitable alternative to others. However, the implementation of continuous production has been hindered by the lytic nature of this expression system and the process-detrimental virus passage effect. In this study, we implemented a multi-stage bioreactor setup for continuous production of influenza hemagglutinin-displaying virus-like particles (HA-VLPs) using IC-BEVS. A setup consisting of one Cell Growth Bioreactor simultaneously feeding non-infected insect cells to three parallel Production Bioreactors operated at different residence times (RT) (18, 36, and 54 h) was implemented; Production Bioreactors were continuously harvested. Two insect cell lines (neutral pH-adapted High Five and Sf9) and two recombinant baculovirus (rBAC) constructs (one that originates from a bacmid, rBAC
bacmid , and another of non-bacteria origin, rBACflashbac ) were tested. Combining rBACflashbac with Sf9 cells was the most efficient approach, allowing consistent HA-VLPs titers (34 ± 14 HA titer/mL) and rBAC titers (108 -109 pfu/mL) throughout the period of continuous operation (20 days). Cell growth kinetics and viability varied across RT, and higher RT was associated with increased expression of HA-VLPs, independent of the cell line and rBAC used; RT of 54 h allowed to maximize titers. The presence of particles resembling HA-VLPs was confirmed by transmission electron microscopy throughout the continuous operation. This work showcases the implementation of a process for continuous production of a promising class of biotherapeutics (i.e., VLPs), and paves the way for establishing continuous, integrated setups using the IC-BEVS expression system., (© 2025 The Author(s). Biotechnology and Bioengineering published by Wiley Periodicals LLC.)- Published
- 2025
- Full Text
- View/download PDF
8. Differential effect of acute versus persistent insect-specific flavivirus infection on superinfection exclusion of West Nile, Zika and chikungunya viruses in RNAi-competent and -deficient mosquito cells.
- Author
-
Willemsen W, Helmes N, Overheul GJ, Henkens M, Spruijt R, van Rij RP, van Oers MM, Pijlman GP, and Fros JJ
- Abstract
Millions of people are annually infected by mosquito-transmitted arboviruses including dengue virus (DENV), West Nile virus (WNV), Zika virus (ZIKV) and chikungunya virus (CHIKV). Insect-specific flaviviruses (ISFs), which only infect mosquitoes and cannot replicate in vertebrates, can offers a potential one health strategy to block the transmission of arboviruses by reducing the mosquito's susceptibility for subsequent arbovirus infections through superinfection exclusion (SIE),. Most SIE studies focus on acute ISF infections in RNAi-deficient Aedes albopictus C6/36 cells. Because ISFs are known to persistently infect mosquitoes, acute infections in C6/36 cells may not accurately reflect natural interactions between ISFs and arboviruses. To study the underlying mechanisms for SIE, we persistently infected C6/36 and RNAi-competent Aedes aegypti Aag2 cells with the ISF Binjari virus (BinJV) and a BinJ-ZIKV chimera that contains the ZIKV prME structural genes. SIE of WNV, ZIKV and CHIKV by BinJV was more pronounced in acute than in persistently infected cells and much stronger in acutely infected C6/36 cells compared to Aag2 cells. The viability of RNAi-deficient mosquito cells was severely reduced upon acute ISF infection, which correlated to the observed SIE. However, persistently infected mosquito cells still inhibited subsequent arbovirus replication. Moreover, RNAi-competent Aag2 cells were better protected against ZIKV superinfection when they were pre-infected with BinJ-ZIKV as compared to BinJV. Therefore, acute ISF infections and strong cytopathic effects in RNAi-deficient cells augment SIE, while in persistently infected cells SIE is established through RNAi-dependent and independent mechanisms. This highlight the importance of using more representative in vitro models., Competing Interests: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (© 2024 The Authors. Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
9. Differential susceptibility of geographically distinct Ixodes ricinus populations to tick-borne encephalitis virus and louping ill virus.
- Author
-
Bakker JW, Esser HJ, Sprong H, Godeke GJ, Hoornweg TE, de Boer WF, Pijlman GP, and Koenraadt CJM
- Subjects
- Animals, Netherlands epidemiology, Austria, Ixodes, Encephalitis Viruses, Tick-Borne genetics, Encephalitis, Tick-Borne epidemiology
- Abstract
Tick-borne encephalitis virus (TBEV) is an emerging pathogen in the Netherlands. Multiple divergent viral strains are circulating and the focal distribution of TBEV remains poorly understood. This may, however, be explained by differences in the susceptibility of tick populations for specific viruses and viral strains, and by viral strains having higher infection success in their local tick population. We investigated this hypothesis by exposing Dutch Ixodes ricinus ticks to two different TBEV strains: TBEV-NL from the Netherlands and TBEV-Neudoerfl from Austria. In addition, we exposed ticks to louping Ill virus (LIV), which is endemic to large parts of the United Kingdom and Ireland, but has not been reported in the Netherlands. Ticks were collected from two locations in the Netherlands: one location without evidence of TBEV circulation and one location endemic for the TBEV-NL strain. Ticks were infected in a biosafety level 3 laboratory using an artificial membrane feeding system. Ticks collected from the region without evidence of TBEV circulation had lower infection rates for TBEV-NL as compared to TBEV-Neudoerfl. Vice versa , ticks collected from the TBEV-NL endemic region had higher infection rates for TBEV-NL compared to TBEV-Neudoerfl. In addition, LIV infection rates were much lower in Dutch ticks compared to TBEV, which may explain why LIV is not present in the Netherlands. Our findings show that ticks from two distinct geographical populations differ in their susceptibility to TBEV strains, which could be the result of differences in the genetic background of the tick populations.
- Published
- 2024
- Full Text
- View/download PDF
10. A getah virus-like-particle vaccine provides complete protection from viremia and arthritis in wild-type mice.
- Author
-
Miao Q, Nguyen W, Zhu J, Liu G, van Oers MM, Tang B, Yan K, Larcher T, Suhrbier A, and Pijlman GP
- Subjects
- Animals, Mice, Alphavirus Infections prevention & control, Alphavirus Infections immunology, Viral Vaccines immunology, Viral Vaccines administration & dosage, Viral Vaccines genetics, Female, Viral Envelope Proteins immunology, Viral Envelope Proteins genetics, Baculoviridae genetics, Baculoviridae immunology, Sf9 Cells, Vaccines, Virus-Like Particle immunology, Vaccines, Virus-Like Particle administration & dosage, Viremia prevention & control, Viremia immunology, Antibodies, Neutralizing immunology, Antibodies, Neutralizing blood, Antibodies, Viral immunology, Antibodies, Viral blood, Mice, Inbred C57BL, Arthritis immunology, Arthritis prevention & control, Alphavirus immunology, Alphavirus genetics
- Abstract
Getah virus (GETV) is an emerging mosquito-borne virus with economic impact on the livestock industry in East Asia. In this study, we successfully produced GETV virus-like particles (VLPs) in insect cells using the baculovirus expression vector system. We show that the GETV envelope glycoproteins were successfully expressed at the surface of the insect cell and were glycosylated. VLPs were isolated from the culture fluid as enveloped particles of 60-80 nm in diameter. Two 1 µg vaccinations with this GETV VLP vaccine, without adjuvant, generated neutralizing antibody responses and protected wild-type C57/BL6 mice against GETV viremia and arthritic disease. The GETV VLP vaccine may find application as a horse and/or pig vaccine in the future., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. Japanese encephalitis virus-induced DNA methylation contributes to blood-brain barrier permeability by modulating tight junction protein expression.
- Author
-
Xiang X, Yu D, Li Z, Fros JJ, Wei J, Liu K, Li Z, Shao D, Li B, Kortekaas J, van Oers MM, Ma Z, Pijlman GP, and Qiu Y
- Subjects
- Animals, Mice, Permeability, Blood-Brain Barrier metabolism, Blood-Brain Barrier virology, DNA Methylation, Encephalitis Virus, Japanese, Tight Junction Proteins metabolism, Tight Junction Proteins genetics, Encephalitis, Japanese metabolism, Encephalitis, Japanese genetics
- Abstract
Japanese encephalitis virus (JEV) is a neurotropic and neuroinvasive flavivirus causing viral encephalitis, which seriously threatens the development of animal husbandry and human health. DNA methylation is a major epigenetic modification involved in viral pathogenesis, yet how DNA methylation affects JEV infection remains unknown. Here, we show genome-wide DNA methylation profiles in the brains of JEV-infected mice compared to mock-infected mice. JEV can significantly increase the overall DNA methylation levels in JEV-infected mouse brains. A total of 14,781 differentially methylated regions associated genes (DMGs) have been identified. Subsequently, KEGG pathway analysis suggested that DNA methylation modulates the tight junction signaling pathway, which can potentially impact the permeability of the blood-brain barrier (BBB). We demonstrate that hypermethylation of the tight junction gene Afdn promoter inhibited AFDN expression and increased monolayer permeability of mouse brain microvascular endothelial (bEnd.3) cells in an in vitro transwell assay. Collectively, this study reveals that DNA methylation is increased in a murine Japanese encephalitis model and that modulation of Afdn expression promotes BBB permeability., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. Packaging of alphavirus-based self-amplifying mRNA yields replication-competent virus through a mechanism of aberrant homologous RNA recombination.
- Author
-
Hick TAH, Geertsema C, Nijland R, and Pijlman GP
- Subjects
- Animals, Humans, Cell Line, Replicon genetics, Virus Assembly, Homologous Recombination, Recombination, Genetic, Virus Replication, Alphavirus genetics, RNA, Viral genetics, RNA, Messenger genetics
- Abstract
Messenger (m)RNA has taken center stage in vaccine development, gene therapy, and cancer immunotherapy. A next-generation of mRNA is the self-amplifying (sa)mRNA, which induces broad and long-lasting immunity at a lower dose which provides better clinical outcomes in conjunction with fewer adverse effects. SamRNA, also known as "replicon" RNA, encodes the replication machinery of an alphavirus together with an antigen. Efficient delivery of replicon RNA to target tissues can be accomplished by packaging the replicon RNA in virus-like replicon particles (VRPs) via co-transfection of producer cells with defective helper RNA(s) encoding the alphavirus structural proteins. During the manufacture of VRPs, however, there is a potential risk of RNA recombination, which may lead to the formation of replication-competent virus (RCV). To investigate the factors influencing the unwanted RCV formation, we evaluated how sequence homology orchestrates alphavirus RNA recombination. Several combinations of complementing alphavirus replicon and helper RNAs varying in length of sequences overlap were co-transfected in mammalian cells. The culture fluid was serially passaged to detect RCV. Nanopore sequencing of cells after the first passage in combination with amplicon-based Sanger sequencing of RCV in the culture fluid after four passages led to the detection of RNA recombination. RCV was generated between replicon and helper RNAs with sequence homology in either the non-structural or structural genes, whereas RNAs without overlapping gene regions did not generate RCV. Remarkably, no sequence overlap was detected at the recombination junction sites in the RCV genome, suggesting a mechanism of "aberrant homologous RNA recombination." Accordingly, we conclude that the alphavirus RNA recombination process leading to the formation of RCV is homology-assisted and can be prevented by avoiding sequence homology between replicon and helper RNAs.IMPORTANCEThere is a growing interest in the use of self-amplifying (sa)mRNA vectors for next-generation vaccine development, gene therapy, and cancer immunotherapy. The delivery of samRNA in the form of virus-like replicon particles (VRPs) enables efficient delivery of samRNA to target tissue. The production of these VRPs, however, suffers from contamination with replication-competent virus (RCV) that is thought to arise from recombination events between samRNA and helper RNAs for VRP packaging. The presence of RCV in samRNA in the clinical product is undesirable as alphaviruses may cause serious disease in humans. However, the underlying recombination mechanism leading to RCV is currently unknown. In our work, we demonstrate a detailed evaluation of the recombination sites, which indicates that RCV is formed through an unusual mechanism of "aberrant homologous RNA recombination." The results are useful for researchers in the field of RNA vaccine manufacture and delivery., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
13. Safety concern of recombination between self-amplifying mRNA vaccines and viruses is mitigated in vivo.
- Author
-
Hick TAH, Geertsema C, Nguyen W, Bishop CR, van Oosten L, Abbo SR, Dumenil T, van Kuppeveld FJM, Langereis MA, Rawle DJ, Tang B, Yan K, van Oers MM, Suhrbier A, and Pijlman GP
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Humans, Receptor, Interferon alpha-beta genetics, Virus Replication, Homeodomain Proteins genetics, Homeodomain Proteins immunology, Vaccines, Synthetic immunology, Vaccines, Synthetic adverse effects, Mice, Knockout, SARS-CoV-2 genetics, SARS-CoV-2 immunology, Viral Vaccines immunology, Viral Vaccines genetics, Viral Vaccines adverse effects, mRNA Vaccines, Alphavirus genetics, Alphavirus immunology, Recombination, Genetic
- Abstract
Self-amplifying mRNA (SAM) vaccines can be rapidly deployed in the event of disease outbreaks. A legitimate safety concern is the potential for recombination between alphavirus-based SAM vaccines and circulating viruses. This theoretical risk needs to be assessed in the regulatory process for SAM vaccine approval. Herein, we undertake extensive in vitro and in vivo assessments to explore recombination between SAM vaccine and a wide selection of alphaviruses and a coronavirus. SAM vaccines were found to effectively limit alphavirus co-infection through superinfection exclusion, although some co-replication was still possible. Using sensitive cell-based assays, replication-competent alphavirus chimeras were generated in vitro as a result of rare, but reproducible, RNA recombination events. The chimeras displayed no increased fitness in cell culture. Viable alphavirus chimeras were not detected in vivo in C57BL/6J, Rag1
-/- and Ifnar-/- mice, in which high levels of SAM vaccine and alphavirus co-replicated in the same tissue. Furthermore, recombination between a SAM-spike vaccine and a swine coronavirus was not observed. In conclusion we state that although the ability of SAM vaccines to recombine with alphaviruses might be viewed as an environmental safety concern, several key factors substantially mitigate against in vivo emergence of chimeric viruses from SAM vaccine recipients., Competing Interests: Declaration of interests The user committee for the RepliSAFE project included members from MSD Animal Health. MSD Animal Health provided TC-83 replicons and antibodies but otherwise provided no other resources or funding, had no input into the manuscript nor the decision to publish., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
14. Subgenomic flavivirus RNA as key target for live-attenuated vaccine development.
- Author
-
Doets K and Pijlman GP
- Subjects
- Humans, Animals, Vaccine Development, Vaccines, Attenuated immunology, Flavivirus immunology, Flavivirus genetics, RNA, Viral genetics, Viral Vaccines immunology, Flavivirus Infections prevention & control, Flavivirus Infections virology
- Abstract
Live-attenuated flavivirus vaccines confer long-term protection against disease, but the design of attenuated flaviviruses does not follow a general approach. The non-coding, subgenomic flavivirus RNA (sfRNA) is produced by all flaviviruses and is an essential factor in viral pathogenesis and transmission. We argue that modulating sfRNA expression is a promising, universal strategy to finetune flavivirus attenuation for developing effective flavivirus vaccines of the future., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
15. Ixodes ricinus as potential vector for Usutu virus.
- Author
-
Bakker JW, Münger E, Esser HJ, Sikkema RS, de Boer WF, Sprong H, Reusken CBEM, de Vries A, Kohl R, van der Linden A, Stroo A, van der Jeugd H, Pijlman GP, Koopmans MPG, Munnink BBO, and Koenraadt CJM
- Subjects
- Animals, Birds virology, Arachnid Vectors virology, Arachnid Vectors physiology, Netherlands, Female, Ixodes virology, Ixodes physiology, Flavivirus physiology, Flavivirus genetics, Flavivirus Infections transmission, Flavivirus Infections veterinary, Flavivirus Infections virology, Nymph virology, Bird Diseases virology, Bird Diseases transmission
- Abstract
Usutu virus (USUV) is an emerging flavivirus that is maintained in an enzootic cycle with mosquitoes as vectors and birds as amplifying hosts. In Europe, the virus has caused mass mortality of wild birds, mainly among Common Blackbird (Turdus merula) populations. While mosquitoes are the primary vectors for USUV, Common Blackbirds and other avian species are exposed to other arthropod ectoparasites, such as ticks. It is unknown, however, if ticks can maintain and transmit USUV. We addressed this question using in vitro and in vivo experiments and field collected data. USUV replicated in IRE/CTVM19 Ixodes ricinus tick cells and in injected ticks. Moreover, I. ricinus nymphs acquired the virus via artificial membrane blood-feeding and maintained the virus for at least 70 days. Transstadial transmission of USUV from nymphs to adults was confirmed in 4.9% of the ticks. USUV disseminated from the midgut to the haemocoel, and was transmitted via the saliva of the tick during artificial membrane blood-feeding. We further explored the role of ticks by monitoring USUV in questing ticks and in ticks feeding on wild birds in the Netherlands between 2016 and 2019. In total, 622 wild birds and the Ixodes ticks they carried were tested for USUV RNA. Of these birds, 48 (7.7%) carried USUV-positive ticks. The presence of negative-sense USUV RNA in ticks, as confirmed via small RNA-sequencing, showed active virus replication. In contrast, we did not detect USUV in 15,381 questing ticks collected in 2017 and 2019. We conclude that I. ricinus can be infected with USUV and can transstadially and horizontally transmit USUV. However, in comparison to mosquito-borne transmission, the role of I. ricinus ticks in the epidemiology of USUV is expected to be minor., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2024 Bakker et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
16. The potential role of the Asian bush mosquito Aedes japonicus as spillover vector for West Nile virus in the Netherlands.
- Author
-
Linthout C, Martins AD, de Wit M, Delecroix C, Abbo SR, Pijlman GP, and Koenraadt CJM
- Subjects
- Animals, Netherlands epidemiology, Humans, Female, Birds virology, Chickens virology, Host-Seeking Behavior, Seasons, Aedes virology, Aedes physiology, Mosquito Vectors virology, Mosquito Vectors physiology, West Nile virus physiology, West Nile Fever transmission, West Nile Fever virology
- Abstract
Background: In recent years the Asian bush mosquito Aedes japonicus has invaded Europe, including the Netherlands. This species is a known vector for a range of arboviruses, possibly including West Nile virus (WNV). As WNV emerged in the Netherlands in 2020, it is important to investigate the vectorial capacity of mosquito species present in the Netherlands to estimate the risk of future outbreaks and further spread of the virus. Therefore, this study evaluates the potential role of Ae. japonicus in WNV transmission and spillover from birds to dead-end hosts in the Netherlands., Methods: We conducted human landing collections in allotment gardens (Lelystad, the Netherlands) in June, August and September 2021 to study the diurnal and seasonal host-seeking behaviour of Ae. japonicus. Furthermore, their host preference in relation to birds using live chicken-baited traps was investigated. Vector competence of field-collected Ae. japonicus mosquitoes for two isolates of WNV at two different temperatures was determined. Based on the data generated from these studies, we developed a Susceptible-Exposed-Infectious-Recovered (SEIR) model to calculate the risk of WNV spillover from birds to humans via Ae. japonicus, under the condition that the virus is introduced and circulates in an enzootic cycle in a given area., Results: Our results show that Ae. japonicus mosquitoes are actively host seeking throughout the day, with peaks in activity in the morning and evening. Their abundance in August was higher than in June and September. For the host-preference experiment, we documented a small number of mosquitoes feeding on birds: only six blood-fed females were caught over 4 full days of sampling. Finally, our vector competence experiments with Ae. japonicus compared to its natural vector Culex pipiens showed a higher infection and transmission rate when infected with a local, Dutch, WNV isolate compared to a Greek isolate of the virus. Interestingly, we also found a small number of infected Cx. pipiens males with virus-positive leg and saliva samples., Conclusions: Combining the field and laboratory derived data, our model predicts that Ae. japonicus could act as a spillover vector for WNV and could be responsible for a high initial invasion risk of WNV when present in large numbers., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
17. Evaluation of bird-adapted self-amplifying mRNA vaccine formulations in chickens.
- Author
-
Comes JDG, Doets K, Zegers T, Kessler M, Slits I, Ballesteros NA, van de Weem NMP, Pouwels H, van Oers MM, van Hulten MCW, Langereis M, and Pijlman GP
- Subjects
- Humans, Animals, Chickens, mRNA Vaccines, Antibodies, Viral, Antibodies, Neutralizing, RNA, Capsid Proteins, Mammals genetics, Viral Vaccines genetics, Poultry Diseases prevention & control
- Abstract
Each year, millions of poultry succumb to highly pathogenic avian influenza A virus (AIV) and infectious bursal disease virus (IBDV) infections. Conventional vaccines based on inactivated or live-attenuated viruses are useful tools for disease prevention and control, yet, they often fall short in terms of safety, efficacy, and development times. Therefore, versatile vaccine platforms are crucial to protect poultry from emerging viral pathogens. Self-amplifying (replicon) RNA vaccines offer a well-defined and scalable option for the protection of both animals and humans. The best-studied replicon platform, based on the Venezuelan equine encephalitis virus (VEEV; family Togaviridae) TC-83 vaccine strain, however, displays limited efficacy in poultry, warranting the exploration of alternative, avian-adapted, replicon platforms. In this study, we engineered two Tembusu virus (TMUV; family Flaviviridae) replicons encoding varying capsid gene lengths and compared these to the benchmark VEEV replicon in vitro. The TMUV replicon system exhibited a robust and prolonged transgene expression compared to the VEEV replicon system in both avian and mammalian cells. Moreover, the TMUV replicon induced a lesser cytopathic effect compared to the VEEV replicon RNA in vitro. DNA-launched versions of the TMUV and VEEV replicons (DREP) were also developed. The replicons successfully expressed the AIV haemagglutinin (HA) glycoproteins and the IBDV capsid protein (pVP2). To assess the immune responses elicited by the TMUV replicon system in chickens, a prime-boost vaccination trial was conducted using lipid nanoparticle (LNP)-formulated replicon RNA and DREP encoding the viral (glyco)proteins of AIV or IBDV. Both TMUV and VEEV replicon RNAs were unable to induce a humoral response against AIV. However, TMUV replicon RNA induced IBDV-specific seroconversion in vaccinated chickens, in contrast to VEEV replicon RNA, which showed no significant humoral response. In both AIV and IBDV immunization studies, VEEV DREP generated the highest (neutralizing) antibody responses, which underscores the potential for self-amplifying mRNA vaccine technology to combat emerging poultry diseases., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Gorben Pijlman reports financial support was provided by MSD Animal Health. Martijn Langereis, Noortje van de Weem, Henk Pouwels, Natalia Ballesteros reports a relationship with MSD Animal Health that includes: employment. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Scarless Baculovirus Genome Editing Using Lambda-Red Recombineering in E. coli.
- Author
-
de Jong LA, van Oosten L, and Pijlman GP
- Subjects
- Chromosomes, Artificial, Bacterial genetics, Genome, Viral, Genetic Engineering methods, Bacteriophage lambda genetics, Homologous Recombination, Gene Editing methods, Escherichia coli genetics, Baculoviridae genetics, Genetic Vectors genetics
- Abstract
Baculoviruses are widely used for their potential as biological pesticide and as platform for the production of recombinant proteins and gene therapy vectors. The Baculovirus Expression Vector System (BEVS) is used for high level of expression of (multiple) proteins in insect cells. Baculovirus recombinants can be quickly constructed by transposition of the gene(s) of interest into a so-called bacmid, which is a baculovirus infectious clone maintained as single-copy, bacterial artificial chromosome in Escherichia coli. A two-step homologous recombineering technique using the lambda-red system in E. coli allows for scarless editing of the bacmid with PCR products based on sequence homology. In the first step, a selection cassette with 50 bp homology arms, typically generated by PCR, is inserted into the designated locus. In the second step, the selection cassette is removed based on a negative selection marker, such as SacB or rpsL. This lambda-red recombineering technique can be used for multiple gene editing purposes, including (large) deletions, insertions, and even single point mutations. Moreover, since there are no remnants of the editing process, successive modifications of the same bacmid are possible. This chapter provides detailed instructions to design and perform two-step homologous recombineering of baculovirus bacmid DNA in E. coli. We present two case studies demonstrating the utility of this technique for creating a deletion mutant of the chitinase and cathepsin genes and for introducing a single point mutation in the baculovirus gene gp41. This scarless genome editing approach can facilitate functional studies of baculovirus genes and improve the production of recombinant proteins using the BEVS., (© 2024. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
19. Infectious clone of a contemporary Tembusu virus and replicons expressing reporter genes or heterologous antigens from poultry viruses.
- Author
-
Comes JDG, Poniman M, van Oosten L, Doets K, de Cloe S, Geertsema C, and Pijlman GP
- Subjects
- Animals, Poultry genetics, Genes, Reporter genetics, DNA, Complementary, Antigens, Heterophile, Chickens, Ducks genetics, Clone Cells, Replicon, Flavivirus Infections veterinary, Flavivirus Infections genetics, Poultry Diseases genetics, Flavivirus genetics, Culicidae
- Abstract
The novel mosquito-borne Tembusu virus (TMUV, family Flaviviridae) was discovered as the cause of a severe outbreak of egg-drop syndrome affecting ducks in Southeast Asia in 2010. TMUV infection can also lead to high mortality in various additional avian species such as geese, pigeons, and chickens. This study describes the construction of an infectious cDNA clone of a contemporary duck-isolate (TMUV WU2016). The virus recovered after transfection of BHK-21 cells shows enhanced virus replication compared to the mosquito-derived MM1775 strain. Next, the WU2016 cDNA clone was modified to create a SP6 promoter-driven, self-amplifying mRNA (replicon) capable of expressing a range of different reporter genes (Renilla luciferase, mScarlet, mCherry, and GFP) and viral (glyco)proteins of avian influenza virus (AIV; family Orthomyxoviridae), infectious bursal disease virus (IDBV; family Bunyaviridae) and infectious bronchitis virus (IBV; family Coronaviridae). The current study demonstrates the flexibility of the TMUV replicon system, to produce different heterologous proteins over an extended period of time and its potential use as a platform technology for novel poultry vaccines., (© 2023 The Authors. Biotechnology Journal published by Wiley-VCH GmbH.)
- Published
- 2024
- Full Text
- View/download PDF
20. Infection of wild-caught wood mice (Apodemus sylvaticus) and yellow-necked mice (A. flavicollis) with tick-borne encephalitis virus.
- Author
-
Bakker JW, Pascoe EL, van de Water S, van Keulen L, de Vries A, Woudstra LC, Esser HJ, Pijlman GP, de Boer WF, Sprong H, Kortekaas J, Wichgers Schreur PJ, and Koenraadt CJM
- Subjects
- Animals, Mice, Murinae, Netherlands, Encephalitis Viruses, Tick-Borne genetics, Encephalitis, Tick-Borne epidemiology, Encephalitis, Tick-Borne veterinary, Ticks
- Abstract
The distribution of tick-borne encephalitis virus (TBEV) is expanding to Western European countries, including the Netherlands, but the contribution of different rodent species to the transmission of TBEV is poorly understood. We investigated whether two species of wild rodents native to the Netherlands, the wood mouse Apodemus sylvaticus and the yellow-necked mouse Apodemus flavicollis, differ in their relative susceptibility to experimental infection with TBEV. Wild-caught individuals were inoculated subcutaneously with the classical European subtype of TBEV (Neudoerfl) or with TBEV-NL, a genetically divergent TBEV strain from the Netherlands. Mice were euthanised and necropsied between 3 and 21 days post-inoculation. None of the mice showed clinical signs or died during the experimental period. Nevertheless, TBEV RNA was detected up to 21 days in the blood of both mouse species and TBEV was also isolated from the brain of some mice. Moreover, no differences in infection rates between virus strains and mouse species were found in blood, spleen, or liver samples. Our results suggest that the wood mouse and the yellow-necked mouse may equally contribute to the transmission cycle of TBEV in the Netherlands. Future experimental infection studies that include feeding ticks will help elucidate the relative importance of viraemic transmission in the epidemiology of TBEV., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
21. Rise of the RNA machines - self-amplification in mRNA vaccine design.
- Author
-
Comes JDG, Pijlman GP, and Hick TAH
- Abstract
mRNA vaccines have won the race for early COVID-19 vaccine approval, yet improvements are necessary to retain this leading role in combating infectious diseases. A next generation of self-amplifying mRNAs, also known as replicons, form an ideal vaccine platform. Replicons induce potent humoral and cellular responses with few adverse effects upon a minimal, single-dose immunization. Delivery of replicons is achieved with virus-like replicon particles (VRPs), or in nonviral vehicles such as liposomes or lipid nanoparticles. Here, we discuss innovative advances, including multivalent, mucosal, and therapeutic replicon vaccines, and highlight novelties in replicon design. As soon as essential safety evaluations have been resolved, this promising vaccine concept can transform into a widely applied clinical platform technology taking center stage in pandemic preparedness., Competing Interests: Declaration of interests No interests are declared., (Copyright © 2023 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
22. The virome of the invasive Asian bush mosquito Aedes japonicus in Europe.
- Author
-
Abbo SR, de Almeida JPP, Olmo RP, Balvers C, Griep JS, Linthout C, Koenraadt CJM, Silva BM, Fros JJ, Aguiar ERGR, Marois E, Pijlman GP, and Marques JT
- Abstract
The Asian bush mosquito Aedes japonicus is rapidly invading North America and Europe. Due to its potential to transmit multiple pathogenic arthropod-borne (arbo)viruses including Zika virus, West Nile virus, and chikungunya virus, it is important to understand the biology of this vector mosquito in more detail. In addition to arboviruses, mosquitoes can also carry insect-specific viruses that are receiving increasing attention due to their potential effects on host physiology and arbovirus transmission. In this study, we characterized the collection of viruses, referred to as the virome, circulating in Ae. japonicus populations in the Netherlands and France. Applying a small RNA-based metagenomic approach to Ae. japonicus , we uncovered a distinct group of viruses present in samples from both the Netherlands and France. These included one known virus, Ae. japonicus narnavirus 1 (AejapNV1), and three new virus species that we named Ae. japonicus totivirus 1 (AejapTV1), Ae. japonicus anphevirus 1 (AejapAV1) and Ae. japonicus bunyavirus 1 (AejapBV1). We also discovered sequences that were presumably derived from two additional novel viruses: Ae. japonicus bunyavirus 2 (AejapBV2) and Ae. japonicus rhabdovirus 1 (AejapRV1). All six viruses induced strong RNA interference responses, including the production of twenty-one nucleotide-sized small interfering RNAs, a signature of active replication in the host. Notably, AejapBV1 and AejapBV2 belong to different viral families; however, no RNA-dependent RNA polymerase sequence has been found for AejapBV2. Intriguingly, our small RNA-based approach identified an ∼1-kb long ambigrammatic RNA that is associated with AejapNV1 as a secondary segment but showed no similarity to any sequence in public databases. We confirmed the presence of AejapNV1 primary and secondary segments, AejapTV1, AejapAV1, and AejapBV1 by reverse transcriptase polymerase chain reaction (PCR) in wild-caught Ae. japonicus mosquitoes. AejapNV1 and AejapTV1 were found at high prevalence (87-100 per cent) in adult females, adult males, and larvae. Using a small RNA-based, sequence-independent metagenomic strategy, we uncovered a conserved and prevalent virome among Ae. japonicus mosquito populations. The high prevalence of AejapNV1 and AejapTV1 across all tested mosquito life stages suggests that these viruses are intimately associated with Ae. japonicus ., Competing Interests: None declared., (© The Author(s) 2023. Published by Oxford University Press.)
- Published
- 2023
- Full Text
- View/download PDF
23. Transcriptomic Profiling Reveals Intense Host-Pathogen Dispute Compromising Homeostasis during Acute Rift Valley Fever Virus Infection.
- Author
-
Bermúdez-Méndez E, Angelino P, van Keulen L, van de Water S, Rockx B, Pijlman GP, Ciuffi A, Kortekaas J, and Wichgers Schreur PJ
- Subjects
- Animals, Sheep, Transcriptome, Liver, Host-Pathogen Interactions, Interferons metabolism, Homeostasis, Rift Valley Fever pathology, Rift Valley fever virus pathogenicity, Low Density Lipoprotein Receptor-Related Protein-1 metabolism
- Abstract
Rift Valley fever virus (RVFV) (family Phenuiviridae ) can cause severe disease, and outbreaks of this mosquito-borne pathogen pose a significant threat to public and animal health. Yet many molecular aspects of RVFV pathogenesis remain incompletely understood. Natural RVFV infections are acute, characterized by a rapid onset of peak viremia during the first days post-infection, followed by a rapid decline. Although in vitro studies identified a major role of interferon (IFN) responses in counteracting the infection, a comprehensive overview of the specific host factors that play a role in RVFV pathogenesis in vivo is still lacking. Here, the host in vivo transcriptional profiles in the liver and spleen tissues of lambs exposed to RVFV are studied using RNA sequencing (RNA-seq) technology. We validate that IFN-mediated pathways are robustly activated in response to infection. We also link the observed hepatocellular necrosis with severely compromised organ function, which is reflected as a marked downregulation of multiple metabolic enzymes essential for homeostasis. Furthermore, we associate the elevated basal expression of LRP1 in the liver with RVFV tissue tropism. Collectively, the results of this study deepen the knowledge of the in vivo host response during RVFV infection and reveal new insights into the gene regulation networks underlying pathogenesis in a natural host. IMPORTANCE Rift Valley fever virus (RVFV) is a mosquito-transmitted pathogen capable of causing severe disease in animals and humans. Outbreaks of RVFV pose a significant threat to public health and can result in substantial economic losses. Little is known about the molecular basis of RVFV pathogenesis in vivo , particularly in its natural hosts. We employed RNA-seq technology to investigate genome-wide host responses in the liver and spleen of lambs during acute RVFV infection. We show that RVFV infection drastically decreases the expression of metabolic enzymes, which impairs normal liver function. Moreover, we highlight that basal expression levels of the host factor LRP1 may be a determinant of RVFV tissue tropism. This study links the typical pathological phenotype induced by RVFV infection with tissue-specific gene expression profiles, thereby improving our understanding of RVFV pathogenesis., Competing Interests: The authors declare no conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
24. Competition between two Usutu virus isolates in cell culture and in the common house mosquito Culex pipiens .
- Author
-
van Bree JWM, Linthout C, van Dijk T, Abbo SR, Fros JJ, Koenraadt CJM, Pijlman GP, and Wang H
- Abstract
Usutu virus (USUV) is a mosquito-borne flavivirus of African origin. Over the past decades, USUV has spread through Europe causing mass die-offs among multiple bird species. The natural transmission cycle of USUV involves Culex spp . mosquitoes as vectors and birds as amplifying hosts. Next to birds and mosquitoes, USUV has also been isolated from multiple mammalian species, including humans, which are considered dead-end hosts. USUV isolates are phylogenetically classified into an African and European branch, subdivided into eight genetic lineages (Africa 1, 2, and 3 and Europe 1, 2, 3, 4, and 5 lineages). Currently, multiple African and European lineages are co-circulating in Europe. Despite increased knowledge of the epidemiology and pathogenicity of the different lineages, the effects of co-infection and transmission efficacy of the co-circulating USUV strains remain unclear. In this study, we report a comparative study between two USUV isolates as follows: a Dutch isolate (USUV-NL, Africa lineage 3) and an Italian isolate (USUV-IT, Europe lineage 2). Upon co-infection, USUV-NL was consistently outcompeted by USUV-IT in mosquito, mammalian, and avian cell lines. In mosquito cells, the fitness advantage of USUV-IT was most prominently observed in comparison to the mammalian or avian cell lines. When Culex pipiens mosquitoes were orally infected with the different isolates, no overall differences in vector competence for USUV-IT and USUV-NL were observed. However, during the in vivo co-infection assay, it was observed that USUV-NL infectivity and transmission were negatively affected by USUV-IT but not vice versa ., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 van Bree, Linthout, van Dijk, Abbo, Fros, Koenraadt, Pijlman and Wang.)
- Published
- 2023
- Full Text
- View/download PDF
25. Novel approaches for the rapid development of rationally designed arbovirus vaccines.
- Author
-
van Bree JWM, Visser I, Duyvestyn JM, Aguilar-Bretones M, Marshall EM, van Hemert MJ, Pijlman GP, van Nierop GP, Kikkert M, Rockx BHG, Miesen P, and Fros JJ
- Abstract
Vector-borne diseases, including those transmitted by mosquitoes, account for more than 17% of infectious diseases worldwide. This number is expected to rise with an increased spread of vector mosquitoes and viruses due to climate change and man-made alterations to ecosystems. Among the most common, medically relevant mosquito-borne infections are those caused by arthropod-borne viruses (arboviruses), especially members of the genera Flavivirus and Alphavirus . Arbovirus infections can cause severe disease in humans, livestock and wildlife. Severe consequences from infections include congenital malformations as well as arthritogenic, haemorrhagic or neuroinvasive disease. Inactivated or live-attenuated vaccines (LAVs) are available for a small number of arboviruses; however there are no licensed vaccines for the majority of these infections. Here we discuss recent developments in pan-arbovirus LAV approaches, from site-directed attenuation strategies targeting conserved determinants of virulence to universal strategies that utilize genome-wide re-coding of viral genomes. In addition to these approaches, we discuss novel strategies targeting mosquito saliva proteins that play an important role in virus transmission and pathogenesis in vertebrate hosts. For rapid pre-clinical evaluations of novel arbovirus vaccine candidates, representative in vitro and in vivo experimental systems are required to assess the desired specific immune responses. Here we discuss promising models to study attenuation of neuroinvasion, neurovirulence and virus transmission, as well as antibody induction and potential for cross-reactivity. Investigating broadly applicable vaccination strategies to target the direct interface of the vertebrate host, the mosquito vector and the viral pathogen is a prime example of a One Health strategy to tackle human and animal diseases., Competing Interests: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (© 2023 The Authors. Published by Elsevier B.V.)
- Published
- 2023
- Full Text
- View/download PDF
26. Comparative Efficacy of Mayaro Virus-Like Particle Vaccines Produced in Insect or Mammalian Cells.
- Author
-
Abbo SR, Nguyen W, Abma-Henkens MHC, van de Kamer D, Savelkoul NHA, Geertsema C, Le TTT, Tang B, Yan K, Dumenil T, van Oers MM, Suhrbier A, and Pijlman GP
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Brazil, Antibodies, Neutralizing, Mammals, Vaccines, Virus-Like Particle genetics, Alphavirus genetics, Chikungunya virus, Aedes, Rheumatic Diseases
- Abstract
Mayaro virus (MAYV) is a mosquito-transmitted alphavirus that causes often debilitating rheumatic disease in tropical Central and South America. There are currently no licensed vaccines or antiviral drugs available for MAYV disease. Here, we generated Mayaro virus-like particles (VLPs) using the scalable baculovirus-insect cell expression system. High-level secretion of MAYV VLPs in the culture fluid of Sf9 insect cells was achieved, and particles with a diameter of 64 to 70 nm were obtained after purification. We characterize a C57BL/6J adult wild-type mouse model of MAYV infection and disease and used this model to compare the immunogenicity of VLPs from insect cells with that of VLPs produced in mammalian cells. Mice received two intramuscular immunizations with 1 μg of nonadjuvanted MAYV VLPs. Potent neutralizing antibody responses were generated against the vaccine strain, BeH407, with comparable activity seen against a contemporary 2018 isolate from Brazil (BR-18), whereas neutralizing activity against chikungunya virus was marginal. Sequencing of BR-18 illustrated that this virus segregates with genotype D isolates, whereas MAYV BeH407 belongs to genotype L. The mammalian cell-derived VLPs induced higher mean neutralizing antibody titers than those produced in insect cells. Both VLP vaccines completely protected adult wild-type mice against viremia, myositis, tendonitis, and joint inflammation after MAYV challenge. IMPORTANCE Mayaro virus (MAYV) is associated with acute rheumatic disease that can be debilitating and can evolve into months of chronic arthralgia. MAYV is believed to have the potential to emerge as a tropical public health threat, especially if it develops the ability to be efficiently transmitted by urban mosquito vectors, such as Aedes aegypti and/or Aedes albopictus. Here, we describe a scalable virus-like particle vaccine against MAYV that induced neutralizing antibodies against a historical and a contemporary isolate of MAYV and protected mice against infection and disease, providing a potential new intervention for MAYV epidemic preparedness.
- Published
- 2023
- Full Text
- View/download PDF
27. Real-time online monitoring of insect cell proliferation and baculovirus infection using digital differential holographic microscopy and machine learning.
- Author
-
Altenburg JJ, Klaverdijk M, Cabosart D, Desmecht L, Brunekreeft-Terlouw SS, Both J, Tegelbeckers VIP, Willekens MLPM, van Oosten L, Hick TAH, van der Aalst TMH, Pijlman GP, van Oers MM, Wijffels RH, and Martens DE
- Subjects
- Animals, Recombinant Proteins metabolism, Insecta, Cell Proliferation, Baculoviridae genetics, Machine Learning, Microscopy, Bioreactors
- Abstract
Real-time, detailed online information on cell cultures is essential for understanding modern biopharmaceutical production processes. The determination of key parameters, such as cell density and viability, is usually based on the offline sampling of bioreactors. Gathering offline samples is invasive, has a low time resolution, and risks altering or contaminating the production process. In contrast, measuring process parameters online provides more safety for the process, has a high time resolution, and thus can aid in timely process control actions. We used online double differential digital holographic microscopy (D3HM) and machine learning to perform non-invasive online cell concentration and viability monitoring of insect cell cultures in bioreactors. The performance of D3HM and the machine learning model was tested for a selected variety of baculovirus constructs, products, and multiplicities of infection (MOI). The results show that with online holographic microscopy insect cell proliferation and baculovirus infection can be monitored effectively in real time with high resolution for a broad range of process parameters and baculovirus constructs. The high-resolution data generated by D3HM showed the exact moment of peak cell densities and temporary events caused by feeding. Furthermore, D3HM allowed us to obtain information on the state of the cell culture at the individual cell level. Combining this detailed, real-time information about cell cultures with methodical machine learning models can increase process understanding, aid in decision-making, and allow for timely process control actions during bioreactor production of recombinant proteins., (© 2022 The Authors. Biotechnology Progress published by Wiley Periodicals LLC on behalf of American Institute of Chemical Engineers.)
- Published
- 2023
- Full Text
- View/download PDF
28. Incomplete bunyavirus particles can cooperatively support virus infection and spread.
- Author
-
Bermúdez-Méndez E, Bronsvoort KF, Zwart MP, van de Water S, Cárdenas-Rey I, Vloet RPM, Koenraadt CJM, Pijlman GP, Kortekaas J, and Wichgers Schreur PJ
- Subjects
- Animals, Humans, Virion metabolism, Mammals, Rift Valley fever virus genetics, Orthobunyavirus, Rift Valley Fever genetics, Rift Valley Fever metabolism, Virus Diseases, Culicidae, Arboviruses
- Abstract
Bunyaviruses lack a specific mechanism to ensure the incorporation of a complete set of genome segments into each virion, explaining the generation of incomplete virus particles lacking one or more genome segments. Such incomplete virus particles, which may represent the majority of particles produced, are generally considered to interfere with virus infection and spread. Using the three-segmented arthropod-borne Rift Valley fever virus as a model bunyavirus, we here show that two distinct incomplete virus particle populations unable to spread autonomously are able to efficiently complement each other in both mammalian and insect cells following co-infection. We further show that complementing incomplete virus particles can co-infect mosquitoes, resulting in the reconstitution of infectious virus that is able to disseminate to the mosquito salivary glands. Computational models of infection dynamics predict that incomplete virus particles can positively impact virus spread over a wide range of conditions, with the strongest effect at intermediate multiplicities of infection. Our findings suggest that incomplete particles may play a significant role in within-host spread and between-host transmission, reminiscent of the infection cycle of multipartite viruses., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2022 Bermúdez-Méndez et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2022
- Full Text
- View/download PDF
29. Zika vector competence data reveals risks of outbreaks: the contribution of the European ZIKAlliance project.
- Author
-
Obadia T, Gutierrez-Bugallo G, Duong V, Nuñez AI, Fernandes RS, Kamgang B, Hery L, Gomard Y, Abbo SR, Jiolle D, Glavinic U, Dupont-Rouzeyrol M, Atyame CM, Pocquet N, Boyer S, Dauga C, Vazeille M, Yébakima A, White MT, Koenraadt CJM, Mavingui P, Vega-Rua A, Veronesi E, Pijlman GP, Paupy C, Busquets N, Lourenço-de-Oliveira R, De Lamballerie X, and Failloux AB
- Subjects
- Animals, Disease Outbreaks, Humans, Infant, Newborn, Mosquito Vectors, Aedes, Zika Virus, Zika Virus Infection
- Abstract
First identified in 1947, Zika virus took roughly 70 years to cause a pandemic unusually associated with virus-induced brain damage in newborns. Zika virus is transmitted by mosquitoes, mainly Aedes aegypti, and secondarily, Aedes albopictus, both colonizing a large strip encompassing tropical and temperate regions. As part of the international project ZIKAlliance initiated in 2016, 50 mosquito populations from six species collected in 12 countries were experimentally infected with different Zika viruses. Here, we show that Ae. aegypti is mainly responsible for Zika virus transmission having the highest susceptibility to viral infections. Other species play a secondary role in transmission while Culex mosquitoes are largely non-susceptible. Zika strain is expected to significantly modulate transmission efficiency with African strains being more likely to cause an outbreak. As the distribution of Ae. aegypti will doubtless expand with climate change and without new marketed vaccines, all the ingredients are in place to relive a new pandemic of Zika., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
30. An S1-Nanoparticle Vaccine Protects against SARS-CoV-2 Challenge in K18-hACE2 Mice.
- Author
-
van Oosten L, Yan K, Rawle DJ, Le TT, Altenburg JJ, Fougeroux C, Goksøyr L, Adriaan de Jongh W, Nielsen MA, Sander AF, Pijlman GP, and Suhrbier A
- Subjects
- Animals, Mice, Mice, Transgenic, SARS-CoV-2, COVID-19 prevention & control, COVID-19 Vaccines standards, Nanoparticles
- Published
- 2022
- Full Text
- View/download PDF
31. Balancing Selection of the Intracellular Pathogen Response in Natural Caenorhabditis elegans Populations.
- Author
-
van Sluijs L, Bosman KJ, Pankok F, Blokhina T, Wilten JIHA, Te Molder DM, Riksen JAG, Snoek BL, Pijlman GP, Kammenga JE, and Sterken MG
- Subjects
- Animals, Biological Evolution, Caenorhabditis elegans genetics, Caenorhabditis elegans immunology, Caenorhabditis elegans virology, Host-Pathogen Interactions genetics, Caenorhabditis elegans Proteins genetics, Nodaviridae pathogenicity
- Abstract
Genetic variation in host populations may lead to differential viral susceptibilities. Here, we investigate the role of natural genetic variation in the Intracellular Pathogen Response (IPR), an important antiviral pathway in the model organism Caenorhabditis elegans against Orsay virus (OrV). The IPR involves transcriptional activity of 80 genes including the pals- genes. We examine the genetic variation in the pals -family for traces of selection and explore the molecular and phenotypic effects of having distinct pals -gene alleles. Genetic analysis of 330 global C. elegans strains reveals that genetic diversity within the IPR-related pals -genes can be categorized in a few haplotypes worldwide. Importantly, two key IPR regulators, pals-22 and pals-25 , are in a genomic region carrying signatures of balancing selection, suggesting that different evolutionary strategies exist in IPR regulation. We infected eleven C. elegans strains that represent three distinct pals-22 pals-25 haplotypes with Orsay virus to determine their susceptibility. For two of these strains, N2 and CB4856, the transcriptional response to infection was also measured. The results indicate that pals-22 pals-25 haplotype shapes the defense against OrV and host genetic variation can result in constitutive activation of IPR genes. Our work presents evidence for balancing genetic selection of immunity genes in C. elegans and provides a novel perspective on the functional diversity that can develop within a main antiviral response in natural host populations., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 van Sluijs, Bosman, Pankok, Blokhina, Wilten, te Molder, Riksen, Snoek, Pijlman, Kammenga and Sterken.)
- Published
- 2022
- Full Text
- View/download PDF
32. Virus infection modulates male sexual behaviour in Caenorhabditis elegans.
- Author
-
van Sluijs L, Liu J, Schrama M, van Hamond S, Vromans SPJM, Scholten MH, Žibrat N, Riksen JAG, Pijlman GP, Sterken MG, and Kammenga JE
- Subjects
- Animals, Caenorhabditis elegans genetics, Humans, Male, Reproduction genetics, Sexual Behavior, Sexual Behavior, Animal, Nodaviridae, Virus Diseases
- Abstract
Mating dynamics follow from natural selection on mate choice and individuals maximizing their reproductive success. Mate discrimination reveals itself by a plethora of behaviours and morphological characteristics, each of which can be affected by pathogens. A key question is how pathogens affect mate choice and outcrossing behaviour. Here we investigated the effect of Orsay virus on the mating dynamics of the androdiecious (male and hermaphrodite) nematode Caenorhabditis elegans. We tested genetically distinct strains and found that viral susceptibility differed between sexes in a genotype-dependent manner with males of reference strain N2 being more resistant than hermaphrodites. Males displayed a constitutively higher expression of intracellular pathogen response (IPR) genes, whereas the antiviral RNAi response did not have increased activity in males. Subsequent monitoring of sex ratios over 10 generations revealed that viral presence can change mating dynamics in isogenic populations. Sexual attraction assays showed that males preferred mating with uninfected rather than infected hermaphrodites. Together our results illustrate for the first time that viral infection can significantly affect male mating choice and suggest altered mating dynamics as a novel cause benefitting outcrossing under pathogenic stress conditions in C. elegans., (© 2021 The Authors. Molecular Ecology published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
33. Two-Component Nanoparticle Vaccine Displaying Glycosylated Spike S1 Domain Induces Neutralizing Antibody Response against SARS-CoV-2 Variants.
- Author
-
van Oosten L, Altenburg JJ, Fougeroux C, Geertsema C, van den End F, Evers WAC, Westphal AH, Lindhoud S, van den Berg W, Swarts DC, Deurhof L, Suhrbier A, Le TT, Torres Morales S, Myeni SK, Kikkert M, Sander AF, de Jongh WA, Dagil R, Nielsen MA, Salanti A, Søgaard M, Keijzer TMP, Weijers D, Eppink MHM, Wijffels RH, van Oers MM, Martens DE, and Pijlman GP
- Subjects
- Animals, COVID-19 immunology, Female, Glycosylation, Mice, Mice, Inbred BALB C, SARS-CoV-2 immunology, Sf9 Cells, Viral Vaccines immunology, Antibodies, Neutralizing metabolism, Nanoparticles metabolism, SARS-CoV-2 metabolism
- Abstract
Vaccines pave the way out of the SARS-CoV-2 pandemic. Besides mRNA and adenoviral vector vaccines, effective protein-based vaccines are needed for immunization against current and emerging variants. We have developed a virus-like particle (VLP)-based vaccine using the baculovirus-insect cell expression system, a robust production platform known for its scalability, low cost, and safety. Baculoviruses were constructed encoding SARS-CoV-2 spike proteins: full-length S, stabilized secreted S, or the S1 domain. Since subunit S only partially protected mice from SARS-CoV-2 challenge, we produced S1 for conjugation to bacteriophage AP205 VLP nanoparticles using tag/catcher technology. The S1 yield in an insect-cell bioreactor was ∼11 mg/liter, and authentic protein folding, efficient glycosylation, partial trimerization, and ACE2 receptor binding was confirmed. Prime-boost immunization of mice with 0.5 μg S1-VLPs showed potent neutralizing antibody responses against Wuhan and UK/B.1.1.7 SARS-CoV-2 variants. This two-component nanoparticle vaccine can now be further developed to help alleviate the burden of COVID-19. IMPORTANCE Vaccination is essential to reduce disease severity and limit the transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Protein-based vaccines are useful to vaccinate the world population and to boost immunity against emerging variants. Their safety profiles, production costs, and vaccine storage temperatures are advantageous compared to mRNA and adenovirus vector vaccines. Here, we use the versatile and scalable baculovirus expression vector system to generate a two-component nanoparticle vaccine to induce potent neutralizing antibody responses against SARS-CoV-2 variants. These nanoparticle vaccines can be quickly adapted as boosters by simply updating the antigen component.
- Published
- 2021
- Full Text
- View/download PDF
34. Heat Stress Reduces the Susceptibility of Caenorhabditis elegans to Orsay Virus Infection.
- Author
-
Huang Y, Sterken MG, van Zwet K, van Sluijs L, Pijlman GP, and Kammenga JE
- Subjects
- Animals, Caenorhabditis elegans genetics, Caenorhabditis elegans immunology, Disease Susceptibility, Genes, Helminth, Caenorhabditis elegans virology, Heat-Shock Response, Nodaviridae pathogenicity, RNA Virus Infections immunology
- Abstract
The nematode Caenorhabditis elegans has been a versatile model for understanding the molecular responses to abiotic stress and pathogens. In particular, the response to heat stress and virus infection has been studied in detail. The Orsay virus (OrV) is a natural virus of C. elegans and infection leads to intracellular infection and proteostatic stress, which activates the intracellular pathogen response (IPR). IPR related gene expression is regulated by the genes pals-22 and pals-25, which also control thermotolerance and immunity against other natural pathogens. So far, we have a limited understanding of the molecular responses upon the combined exposure to heat stress and virus infection. We test the hypothesis that the response of C. elegans to OrV infection and heat stress are co-regulated and may affect each other. We conducted a combined heat-stress-virus infection assay and found that after applying heat stress, the susceptibility of C. elegans to OrV was decreased. This difference was found across different wild types of C. elegans . Transcriptome analysis revealed a list of potential candidate genes associated with heat stress and OrV infection. Subsequent mutant screens suggest that pals-22 provides a link between viral response and heat stress, leading to enhanced OrV tolerance of C. elegans after heat stress.
- Published
- 2021
- Full Text
- View/download PDF
35. Punctuated Loci on Chromosome IV Determine Natural Variation in Orsay Virus Susceptibility of Caenorhabditis elegans Strains Bristol N2 and Hawaiian CB4856.
- Author
-
Sterken MG, van Sluijs L, Wang YA, Ritmahan W, Gultom ML, Riksen JAG, Volkers RJM, Snoek LB, Pijlman GP, and Kammenga JE
- Subjects
- Animals, Genes, Helminth, Genetic Predisposition to Disease, Host-Pathogen Interactions, Multifactorial Inheritance, Nodaviridae physiology, Polymorphism, Single Nucleotide, Viral Load, Caenorhabditis elegans genetics, Caenorhabditis elegans virology, Caenorhabditis elegans Proteins genetics, Chromosomes genetics, Cullin Proteins genetics, Genetic Variation, Nodaviridae pathogenicity, Quantitative Trait Loci
- Abstract
Host-pathogen interactions play a major role in evolutionary selection and shape natural genetic variation. The genetically distinct Caenorhabditis elegans strains, Bristol N2 and Hawaiian CB4856, are differentially susceptible to the Orsay virus (OrV). Here, we report the dissection of the genetic architecture of susceptibility to OrV infection. We compare OrV infection in the relatively resistant wild-type CB4856 strain to the more susceptible canonical N2 strain. To gain insight into the genetic architecture of viral susceptibility, 52 fully sequenced recombinant inbred lines (CB4856 × N2 RILs) were exposed to OrV. This led to the identification of two loci on chromosome IV associated with OrV resistance. To verify the two loci and gain additional insight into the genetic architecture controlling virus infection, introgression lines (ILs) that together cover chromosome IV, were exposed to OrV. Of the 27 ILs used, 17 had an CB4856 introgression in an N2 background, and 10 had an N2 introgression in a CB4856 background. Infection of the ILs confirmed and fine-mapped the locus underlying variation in OrV susceptibility, and we found that a single nucleotide polymorphism in cul-6 may contribute to the difference in OrV susceptibility between N2 and CB4856. An allele swap experiment showed the strain CB4856 became as susceptible as the N2 strain by having an N2 cul-6 allele, although having the CB4856 cul-6 allele did not increase resistance in N2. In addition, we found that multiple strains with nonoverlapping introgressions showed a distinct infection phenotype from the parental strain, indicating that there are punctuated locations on chromosome IV determining OrV susceptibility. Thus, our findings reveal the genetic complexity of OrV susceptibility in C. elegans and suggest that viral susceptibility is governed by multiple genes. IMPORTANCE Genetic variation determines the viral susceptibility of hosts. Yet, pinpointing which genetic variants determine viral susceptibility remains challenging. Here, we have exploited the genetic tractability of the model organism Caenorhabditis elegans to dissect the genetic architecture of Orsay virus infection. Our results provide novel insight into natural determinants of Orsay virus infection., (Copyright © 2021 Sterken et al.)
- Published
- 2021
- Full Text
- View/download PDF
36. The dinucleotide composition of the Zika virus genome is shaped by conflicting evolutionary pressures in mammalian hosts and mosquito vectors.
- Author
-
Fros JJ, Visser I, Tang B, Yan K, Nakayama E, Visser TM, Koenraadt CJM, van Oers MM, Pijlman GP, Suhrbier A, and Simmonds P
- Subjects
- A549 Cells, Aedes virology, Animals, Base Composition physiology, Base Sequence genetics, Cell Line, Chlorocebus aethiops, CpG Islands physiology, Dinucleoside Phosphates analysis, Dinucleoside Phosphates genetics, Host Adaptation genetics, Humans, Male, Mammals virology, Mice, Mice, Inbred C57BL, Mice, Knockout, Mosquito Vectors genetics, Mosquito Vectors virology, RNA, Viral chemistry, RNA, Viral genetics, Selection, Genetic physiology, Vero Cells, Zika Virus Infection genetics, Zika Virus Infection transmission, Zika Virus Infection virology, Evolution, Molecular, Genome, Viral genetics, Host-Pathogen Interactions genetics, Zika Virus genetics
- Abstract
Most vertebrate RNA viruses show pervasive suppression of CpG and UpA dinucleotides, closely resembling the dinucleotide composition of host cell transcriptomes. In contrast, CpG suppression is absent in both invertebrate mRNA and RNA viruses that exclusively infect arthropods. Arthropod-borne (arbo) viruses are transmitted between vertebrate hosts by invertebrate vectors and thus encounter potentially conflicting evolutionary pressures in the different cytoplasmic environments. Using a newly developed Zika virus (ZIKV) model, we have investigated how demands for CpG suppression in vertebrate cells can be reconciled with potentially quite different compositional requirements in invertebrates and how this affects ZIKV replication and transmission. Mutant viruses with synonymously elevated CpG or UpA dinucleotide frequencies showed attenuated replication in vertebrate cell lines, which was rescued by knockout of the zinc-finger antiviral protein (ZAP). Conversely, in mosquito cells, ZIKV mutants with elevated CpG dinucleotide frequencies showed substantially enhanced replication compared to wild type. Host-driven effects on virus replication attenuation and enhancement were even more apparent in mouse and mosquito models. Infections with CpG- or UpA-high ZIKV mutants in mice did not cause typical ZIKV-induced tissue damage and completely protected mice during subsequent challenge with wild-type virus, which demonstrates their potential as live-attenuated vaccines. In contrast, the CpG-high mutants displayed enhanced replication in Aedes aegypti mosquitoes and a larger proportion of mosquitoes carried infectious virus in their saliva. These findings show that mosquito cells are also capable of discriminating RNA based on dinucleotide composition. However, the evolutionary pressure on the CpG dinucleotides of viral genomes in arthropod vectors directly opposes the pressure present in vertebrate host cells, which provides evidence that an adaptive compromise is required for arbovirus transmission. This suggests that the genome composition of arbo flaviviruses is crucial to maintain the balance between high-level replication in the vertebrate host and persistent replication in the mosquito vector., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
37. Effect of blood source on vector competence of Culex pipiens biotypes for Usutu virus.
- Author
-
Abbo SR, Visser TM, Koenraadt CJM, Pijlman GP, and Wang H
- Subjects
- Animals, Blood virology, Chickens virology, Culex virology, Feeding Behavior, Flavivirus genetics, Flavivirus isolation & purification, Flavivirus Infections blood, Flavivirus Infections transmission, Humans, Mosquito Vectors virology, Poultry Diseases blood, Poultry Diseases transmission, Viral Zoonoses transmission, Viral Zoonoses virology, Culex physiology, Flavivirus physiology, Flavivirus Infections veterinary, Flavivirus Infections virology, Mosquito Vectors physiology, Poultry Diseases virology
- Abstract
Background: Infectious blood meal experiments have been frequently performed with different virus-vector combinations to assess the transmission potential of arthropod-borne (arbo)viruses. A wide variety of host blood sources have been used to deliver arboviruses to their arthropod vectors in laboratory studies. The type of blood used during vector competence experiments does not always reflect the blood from the viremic vertebrate hosts in the field, but little is known about the effect of blood source on the experimental outcome of vector competence studies. Here we investigated the effect of avian versus human blood on the infection and transmission rates of the zoonotic Usutu virus (USUV) in its primary mosquito vector Culex pipiens., Methods: Cx. pipiens biotypes (pipiens and molestus) were orally infected with USUV through infectious blood meals containing either chicken or human whole blood. The USUV infection and transmission rates were determined by checking mosquito bodies and saliva for USUV presence after 14 days of incubation at 28 °C. In addition, viral titers were determined for USUV-positive mosquito bodies and saliva., Results: Human and chicken blood lead to similar USUV transmission rates for Cx. pipiens biotype pipiens (18% and 15%, respectively), while human blood moderately but not significantly increased the transmission rate (30%) compared to chicken blood (17%) for biotype molestus. USUV infection rates with human blood were consistently higher in both Cx. pipiens biotypes compared to chicken blood. In virus-positive mosquitoes, USUV body and saliva titers did not differ between mosquitoes taking either human or chicken blood. Importantly, biotype molestus had much lower USUV saliva titers compared to biotype pipiens, regardless of which blood was offered., Conclusions: Infection of mosquitoes with human blood led to higher USUV infection rates as compared to chicken blood. However, the blood source had no effect on the vector competence for USUV. Interestingly, biotype molestus is less likely to transmit USUV compared to biotype pipiens due to very low virus titers in the saliva.
- Published
- 2021
- Full Text
- View/download PDF
38. Insect-Specific Flavivirus Replication in Mammalian Cells Is Inhibited by Physiological Temperature and the Zinc-Finger Antiviral Protein.
- Author
-
Colmant AMG, Hobson-Peters J, Slijkerman TAP, Harrison JJ, Pijlman GP, van Oers MM, Simmonds P, Hall RA, and Fros JJ
- Subjects
- A549 Cells, Aedes cytology, Animals, Cell Line, Chlorocebus aethiops, Flavivirus classification, Gene Knockout Techniques, Genome, Viral, Humans, Vero Cells, Aedes virology, Flavivirus genetics, Flavivirus physiology, RNA-Binding Proteins genetics, Temperature, Virus Replication genetics
- Abstract
The genus Flavivirus contains pathogenic vertebrate-infecting flaviviruses (VIFs) and insect-specific flaviviruses (ISF). ISF transmission to vertebrates is inhibited at multiple stages of the cellular infection cycle, via yet to be elucidated specific antiviral responses. The zinc-finger antiviral protein (ZAP) in vertebrate cells can bind CpG dinucleotides in viral RNA, limiting virus replication. Interestingly, the genomes of ISFs contain more CpG dinucleotides compared to VIFs. In this study, we investigated whether ZAP prevents two recently discovered lineage II ISFs, Binjari (BinJV) and Hidden Valley viruses (HVV) from replicating in vertebrate cells. BinJV protein and dsRNA replication intermediates were readily observed in human ZAP knockout cells when cultured at 34 °C. In ZAP-expressing cells, inhibition of the interferon response via interferon response factors 3/7 did not improve BinJV protein expression, whereas treatment with kinase inhibitor C16, known to reduce ZAP's antiviral function, did. Importantly, at 34 °C, both BinJV and HVV successfully completed the infection cycle in human ZAP knockout cells evident from infectious progeny virus in the cell culture supernatant. Therefore, we identify vertebrate ZAP as an important barrier that protects vertebrate cells from ISF infection. This provides new insights into flavivirus evolution and the mechanisms associated with host switching.
- Published
- 2021
- Full Text
- View/download PDF
39. Caprine MAVS Is a RIG-I Interacting Type I Interferon Inducer Downregulated by Peste des Petits Ruminants Virus Infection.
- Author
-
Miao Q, Qi R, Meng C, Zhu J, Tang A, Dong D, Guo H, van Oers MM, Pijlman GP, and Liu G
- Subjects
- Animals, Chlorocebus aethiops, Epithelial Cells, Goats, HEK293 Cells, Humans, Interferon Type I immunology, Vero Cells, Adaptor Proteins, Signal Transducing immunology, Interferon Inducers immunology, Peste-des-Petits-Ruminants immunology, Peste-des-petits-ruminants virus immunology
- Abstract
The mitochondrial antiviral-signaling protein (MAVS, also known as VISA, IPS-1, or CARDIF) plays an essential role in the type I interferon (IFN) response and in retinoic acid-inducible gene I (RIG-I) mediated antiviral innate immunity in mammals. In this study, the caprine MAVS gene (caMAVS, 1566 bp) was identified and cloned. The caMAVS shares the highest amino acid similarity (98.1%) with the predicted sheep MAVS. Confocal microscopy analysis of partial deletion mutants of caMAVS revealed that the transmembrane and the so-called Non-Characterized domains are indispensable for intracellular localization to mitochondria. Overexpression of caMAVS in caprine endometrial epithelial cells up-regulated the mRNA levels of caprine interferon-stimulated genes. We concluded that caprine MAVS mediates the activation of the type I IFN pathway. We further demonstrated that both the CARD-like domain and the transmembrane domain of caMAVS were essential for the activation of the IFN-β promotor. The interaction between caMAVS and caprine RIG-I and the vital role of the CARD and NC domain in this interaction was demonstrated by co-immunoprecipitation. Upon infection with the Peste des Petits Ruminants Virus (PPRV, genus Morbillivirus), the level of MAVS was greatly reduced. This reduction was prevented by the addition of the proteasome inhibitor MG132. Moreover, we found that viral protein V could interact and colocalize with MAVS. Together, we identified caMAVS as a RIG-I interactive protein involved in the activation of type I IFN pathways in caprine cells and as a target for PPRV immune evasion.
- Published
- 2021
- Full Text
- View/download PDF
40. A Tale of 20 Alphaviruses; Inter-species Diversity and Conserved Interactions Between Viral Non-structural Protein 3 and Stress Granule Proteins.
- Author
-
Nowee G, Bakker JW, Geertsema C, Ros VID, Göertz GP, Fros JJ, and Pijlman GP
- Abstract
Alphaviruses infect a diverse range of host organisms including mosquitoes, mammals, and birds. The enigmatic alphavirus non-structural protein 3 (nsP3) has an intrinsically disordered, C-terminal hypervariable domain (HVD) that can interact with a variety of host proteins associated with stress granules (SGs). The HVD displays the highest variability across the more than 30 known alphaviruses, yet it also contains several motifs that are conserved amongst different subgroups of alphaviruses. For some alphaviruses, specific nsP3-SG protein interactions are essential for virus replication. However, it remains difficult to attribute general roles to these virus-host interactions, as multiple amino acid motifs in the HDV display a degree of redundancy and previous studies were performed with a limited number of alphaviruses. To better understand nsP3-host protein interactions we conducted comprehensive co-localization experiments with the nsP3s of 20 diverse alphaviruses: chikungunya, Semliki Forest, Sindbis, Bebaru, Barmah Forest, Getah, Mayaro, Middelburg, O'nyong-nyong, Ross River QML and T48, Una, Whataroa, Southern Elephant Seal, Eilat, Tai Forest (TAFV), Venezuelan/Eastern/Western equine encephalitis (V/E/WEEV) and the aquatic Salmonid alphavirus (SAV), with three different SG proteins (G3BP and its insect homolog Rasputin, FMRP) and BIN1 in mammalian and mosquito cell lines. Despite that all terrestrial alphavirus nsP3s contained at least one BIN1-binding motif (PxPxPR), not all nsP3s co-localized with BIN1. Further, all alphaviruses except SAV, TAFV and VEEV displayed co-localization with G3BP. Although viruses lacking FGxF-like motifs contained Agenet-like domain binding motifs to facilitate interaction with FMRP, cytoplasmic nsP3 granules of all tested alphaviruses co-localized with FMRP. Crispr-Cas9 knockout of G3BP in mammalian cells abolished nsP3-FMRP co-localization for all alphaviruses except V/E/WEEV nsP3s that bind FMRP directly. G3BP knockout also changed nsP3 subcellular localization of Bebaru, Barmah Forest, Getah, and Sindbis viruses. Taken together this study paints a more detailed picture of the diverse interactions between alphavirus nsP3 and SG-associated host proteins. The interaction between nsP3 and G3BP clearly plays a central role and results in recruitment of additional host proteins such as FMRP. However, direct binding of FMRP can make the interaction with G3BP redundant which exemplifies the alternate evolutionary paths of alphavirus subgroups., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Nowee, Bakker, Geertsema, Ros, Göertz, Fros and Pijlman.)
- Published
- 2021
- Full Text
- View/download PDF
41. Relocation of the attTn7 Transgene Insertion Site in Bacmid DNA Enhances Baculovirus Genome Stability and Recombinant Protein Expression in Insect Cells.
- Author
-
Pijlman GP, Grose C, Hick TAH, Breukink HE, van den Braak R, Abbo SR, Geertsema C, van Oers MM, Martens DE, and Esposito D
- Subjects
- Animals, Bioreactors, Cell Line, Chikungunya virus immunology, Genetic Engineering, Genetic Vectors genetics, Insecta, Sf9 Cells, Vaccines, Virus-Like Particle immunology, Baculoviridae genetics, Genome, Viral, Genomic Instability, Homologous Recombination, Mutagenesis, Insertional, Recombinant Proteins genetics, Transgenes
- Abstract
Baculovirus expression vectors are successfully used for the commercial production of complex (glyco)proteins in eukaryotic cells. The genome engineering of single-copy baculovirus infectious clones (bacmids) in E. coli has been valuable in the study of baculovirus biology, but bacmids are not yet widely applied as expression vectors. An important limitation of first-generation bacmids for large-scale protein production is the rapid loss of gene of interest (GOI) expression. The instability is caused by the mini-F replicon in the bacmid backbone, which is non-essential for baculovirus replication in insect cells, and carries the adjacent GOI in between attTn7 transposition sites. In this paper, we test the hypothesis that relocation of the attTn7 transgene insertion site away from the mini-F replicon prevents deletion of the GOI, thereby resulting in higher and prolonged recombinant protein expression levels. We applied lambda red genome engineering combined with SacB counterselection to generate a series of bacmids with relocated attTn7 sites and tested their performance by comparing the relative expression levels of different GOIs. We conclude that GOI expression from the odv-e56 ( pif-5 ) locus results in higher overall expression levels and is more stable over serial passages compared to the original bacmid. Finally, we evaluated this improved next-generation bacmid during a bioreactor scale-up of Sf9 insect cells in suspension to produce enveloped chikungunya virus-like particles as a model vaccine.
- Published
- 2020
- Full Text
- View/download PDF
42. Competition between Usutu virus and West Nile virus during simultaneous and sequential infection of Culex pipiens mosquitoes.
- Author
-
Wang H, Abbo SR, Visser TM, Westenberg M, Geertsema C, Fros JJ, Koenraadt CJM, and Pijlman GP
- Subjects
- Animals, Cell Line, Chlorocebus aethiops, Flavivirus Infections virology, Insect Vectors virology, Mosquito Vectors virology, Vero Cells, Viral Load, Virus Replication, West Nile Fever transmission, West Nile Fever virology, Coinfection virology, Culex virology, Flavivirus physiology, Flavivirus Infections transmission, West Nile virus physiology
- Abstract
Usutu virus (USUV) and West Nile virus (WNV) are closely related mosquito-borne flaviviruses that are mainly transmitted between bird hosts by vector mosquitoes. Infections in humans are incidental but can cause severe disease. USUV is endemic in large parts of Europe, while WNV mainly circulates in Southern Europe. In recent years, WNV is also frequently detected in Northern Europe, thereby expanding the area where both viruses co-circulate. However, it remains unclear how USUV may affect the future spread of WNV and the likelihood of human co-infection. Here we investigated whether co-infections with both viruses in cell lines and their primary mosquito vector, Culex pipiens , affect virus replication and transmission dynamics. We show that USUV is outcompeted by WNV in mammalian, avian and mosquito cells during co-infection. Mosquitoes that were exposed to both viruses simultaneously via infectious blood meal displayed significantly reduced USUV transmission compared to mosquitoes that were only exposed to USUV (from 15% to 3%), while the infection and transmission of WNV was unaffected. In contrast, when mosquitoes were pre-infected with USUV via infectious blood meal, WNV transmission was significantly reduced (from 44% to 17%). Injection experiments established the involvement of the midgut in the observed USUV-mediated WNV inhibition. The competition between USUV and WNV during co-infection clearly indicates that the chance of concurrent USUV and WNV transmission via a single mosquito bite is low. The competitive relation between USUV and WNV may impact virus transmission dynamics in the field and affect the epidemiology of WNV in Europe.
- Published
- 2020
- Full Text
- View/download PDF
43. Experimental adaptation of dengue virus 1 to Aedes albopictus mosquitoes by in vivo selection.
- Author
-
Bellone R, Lequime S, Jupille H, Göertz GP, Aubry F, Mousson L, Piorkowski G, Yen PS, Gabiane G, Vazeille M, Sakuntabhai A, Pijlman GP, de Lamballerie X, Lambrechts L, and Failloux AB
- Subjects
- Animals, Dengue epidemiology, Dengue transmission, Epistasis, Genetic, Humans, Adaptation, Physiological, Aedes virology, Dengue Virus physiology, Mosquito Vectors virology
- Abstract
In most of the world, Dengue virus (DENV) is mainly transmitted by the mosquito Aedes aegypti while in Europe, Aedes albopictus is responsible for human DENV cases since 2010. Identifying mutations that make DENV more competent for transmission by Ae. albopictus will help to predict emergence of epidemic strains. Ten serial passages in vivo in Ae. albopictus led to select DENV-1 strains with greater infectivity for this vector in vivo and in cultured mosquito cells. These changes were mediated by multiple adaptive mutations in the virus genome, including a mutation at position 10,418 in the DENV 3'UTR within an RNA stem-loop structure involved in subgenomic flavivirus RNA production. Using reverse genetics, we showed that the 10,418 mutation alone does not confer a detectable increase in transmission efficiency in vivo. These results reveal the complex adaptive landscape of DENV transmission by mosquitoes and emphasize the role of epistasis in shaping evolutionary trajectories of DENV variants.
- Published
- 2020
- Full Text
- View/download PDF
44. Impact of Gut Bacteria on the Infection and Transmission of Pathogenic Arboviruses by Biting Midges and Mosquitoes.
- Author
-
Möhlmann TWR, Vogels CBF, Göertz GP, Pijlman GP, Ter Braak CJF, Te Beest DE, Hendriks M, Nijhuis EH, Warris S, Drolet BS, van Overbeek L, and Koenraadt CJM
- Subjects
- Animals, Bacterial Physiological Phenomena, Female, Mosquito Vectors virology, Aedes virology, Ceratopogonidae virology, Chikungunya virus physiology, Gastrointestinal Microbiome physiology, Insect Vectors virology, Orthobunyavirus physiology, Zika Virus physiology
- Abstract
Tripartite interactions among insect vectors, midgut bacteria, and viruses may determine the ability of insects to transmit pathogenic arboviruses. Here, we investigated the impact of gut bacteria on the susceptibility of Culicoides nubeculosus and Culicoides sonorensis biting midges for Schmallenberg virus, and of Aedes aegypti mosquitoes for Zika and chikungunya viruses. Gut bacteria were manipulated by treating the adult insects with antibiotics. The gut bacterial communities were investigated using Illumina MiSeq sequencing of 16S rRNA, and susceptibility to arbovirus infection was tested by feeding insects with an infectious blood meal. Antibiotic treatment led to changes in gut bacteria for all insects. Interestingly, the gut bacterial composition of untreated Ae. aegypti and C. nubeculosus showed Asaia as the dominant genus, which was drastically reduced after antibiotic treatment. Furthermore, antibiotic treatment resulted in relatively more Delftia bacteria in both biting midge species, but not in mosquitoes. Antibiotic treatment and subsequent changes in gut bacterial communities were associated with a significant, 1.8-fold increased infection rate of C. nubeculosus with Schmallenberg virus, but not for C. sonorensis. We did not find any changes in infection rates for Ae. aegypti mosquitoes with Zika or chikungunya virus. We conclude that resident gut bacteria may dampen arbovirus transmission in biting midges, but not so in mosquitoes. Use of antimicrobial compounds at livestock farms might therefore have an unexpected contradictory effect on the health of animals, by increasing the transmission of viral pathogens by biting midges.
- Published
- 2020
- Full Text
- View/download PDF
45. Forced Zika Virus Infection of Culex pipiens Leads to Limited Virus Accumulation in Mosquito Saliva.
- Author
-
Abbo SR, Vogels CBF, Visser TM, Geertsema C, van Oers MM, Koenraadt CJM, and Pijlman GP
- Subjects
- Animals, Cell Line, Chlorocebus aethiops, Flavivirus growth & development, Injections, Mosquito Vectors virology, Netherlands, Species Specificity, Vero Cells, Zika Virus Infection transmission, Culex virology, Saliva virology, Zika Virus growth & development, Zika Virus isolation & purification
- Abstract
Zika virus (ZIKV) is a mosquito-borne pathogen that caused a large outbreak in the Americas in 2015 and 2016. The virus is currently present in tropical areas around the globe and can cause severe disease in humans, including Guillain-Barré syndrome and congenital microcephaly. The tropical yellow fever mosquito, Aedes aegypti , is the main vector in the urban transmission cycles of ZIKV. The discovery of ZIKV in wild-caught Culex mosquitoes and the ability of Culex quinquefasciatus mosquitoes to transmit ZIKV in the laboratory raised the question of whether the common house mosquito Culex pipiens , which is abundantly present in temperate regions in North America, Asia and Europe, could also be involved in ZIKV transmission. In this study, we investigated the vector competence of Cx. pipiens (biotypes molestus and pipiens ) from the Netherlands for ZIKV, using Usutu virus as a control. After an infectious blood meal containing ZIKV, none of the tested mosquitoes accumulated ZIKV in the saliva, although 2% of the Cx. pipiens pipiens mosquitoes showed ZIKV-positive bodies. To test the barrier function of the mosquito midgut on virus transmission, ZIKV was forced into Cx. pipiens mosquitoes by intrathoracic injection, resulting in 74% ( molestus ) and 78% ( pipiens ) ZIKV-positive bodies. Strikingly, 14% ( molestus ) and 7% ( pipiens ) of the tested mosquitoes accumulated ZIKV in the saliva after injection. This is the first demonstration of ZIKV accumulation in the saliva of Cx. pipiens upon forced infection. Nevertheless, a strong midgut barrier restricted virus dissemination in the mosquito after oral exposure and we, therefore, consider Cx. pipiens as a highly inefficient vector for ZIKV.
- Published
- 2020
- Full Text
- View/download PDF
46. Reverse Genetics System for Shuni Virus, an Emerging Orthobunyavirus with Zoonotic Potential.
- Author
-
Oymans J, Wichgers Schreur PJ, van Oort S, Vloet R, Venter M, Pijlman GP, van Oers MM, and Kortekaas J
- Subjects
- Animals, Bunyaviridae Infections transmission, Communicable Diseases, Emerging transmission, Genome, Viral, High-Throughput Nucleotide Sequencing, Mice, Open Reading Frames, Orthobunyavirus classification, Phylogeny, RNA, Viral, Rats, United Kingdom epidemiology, Viral Zoonoses transmission, Bunyaviridae Infections epidemiology, Bunyaviridae Infections virology, Communicable Diseases, Emerging epidemiology, Communicable Diseases, Emerging virology, Orthobunyavirus genetics, Reverse Genetics, Viral Zoonoses epidemiology, Viral Zoonoses virology
- Abstract
The genus Orthobunyavirus (family Peribunyaviridae , order Bunyavirales ) comprises over 170 named mosquito- and midge-borne viruses, several of which cause severe disease in animals or humans. Their three-segmented genomes enable reassortment with related viruses, which may result in novel viruses with altered host or tissue tropism and virulence. One such reassortant, Schmallenberg virus (SBV), emerged in north-western Europe in 2011. Shuni virus (SHUV) is an orthobunyavirus related to SBV that is associated with neurological disease in horses in southern Africa and recently caused an outbreak manifesting with neurological disease and birth defects among ruminants in Israel. The zoonotic potential of SHUV was recently underscored by its association with neurological disease in humans. We here report a reverse genetics system for SHUV and provide first evidence that the non-structural (NSs) protein of SHUV functions as an antagonist of host innate immune responses. We furthermore report the rescue of a reassortant containing the L and S segments of SBV and the M segment of SHUV. This novel reverse genetics system can now be used to study SHUV virulence and tropism, and to elucidate the molecular mechanisms that drive reassortment events.
- Published
- 2020
- Full Text
- View/download PDF
47. The invasive Asian bush mosquito Aedes japonicus found in the Netherlands can experimentally transmit Zika virus and Usutu virus.
- Author
-
Abbo SR, Visser TM, Wang H, Göertz GP, Fros JJ, Abma-Henkens MHC, Geertsema C, Vogels CBF, Koopmans MPG, Reusken CBEM, Hall-Mendelin S, Hall RA, van Oers MM, Koenraadt CJM, and Pijlman GP
- Subjects
- Animals, Female, Flavivirus, Humans, Microcephaly virology, Netherlands, Saliva virology, Temperature, Zika Virus, Aedes virology, Flavivirus Infections transmission, Mosquito Vectors virology, Zika Virus Infection transmission
- Abstract
Background: The Asian bush mosquito Aedes japonicus is invading Europe and was first discovered in Lelystad, the Netherlands in 2013, where it has established a permanent population. In this study, we investigated the vector competence of Ae. japonicus from the Netherlands for the emerging Zika virus (ZIKV) and zoonotic Usutu virus (USUV). ZIKV causes severe congenital microcephaly and Guillain-Barré syndrome in humans. USUV is closely related to West Nile virus, has recently spread throughout Europe and is causing mass mortality of birds. USUV infection in humans can result in clinical manifestations ranging from mild disease to severe neurological impairments., Methodology/principal Findings: In our study, field-collected Ae. japonicus females received an infectious blood meal with ZIKV or USUV by droplet feeding. After 14 days at 28°C, 3% of the ZIKV-blood fed mosquitoes and 13% of the USUV-blood fed mosquitoes showed virus-positive saliva, indicating that Ae. japonicus can transmit both viruses. To investigate the effect of the mosquito midgut barrier on virus transmission, female mosquitoes were intrathoracically injected with ZIKV or USUV. Of the injected mosquitoes, 96% (ZIKV) and 88% (USUV) showed virus-positive saliva after 14 days at 28°C. This indicates that ZIKV and USUV can efficiently replicate in Ae. japonicus but that a strong midgut barrier is normally restricting virus dissemination. Small RNA deep sequencing of orally infected mosquitoes confirmed active replication of ZIKV and USUV, as demonstrated by potent small interfering RNA responses against both viruses. Additionally, de novo small RNA assembly revealed the presence of a novel narnavirus in Ae. japonicus., Conclusions/significance: Given that Ae. japonicus can experimentally transmit arthropod-borne viruses (arboviruses) like ZIKV and USUV and is currently expanding its territories, we should consider this mosquito as a potential vector for arboviral diseases in Europe., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
48. Subgenomic flavivirus RNA binds the mosquito DEAD/H-box helicase ME31B and determines Zika virus transmission by Aedes aegypti .
- Author
-
Göertz GP, van Bree JWM, Hiralal A, Fernhout BM, Steffens C, Boeren S, Visser TM, Vogels CBF, Abbo SR, Fros JJ, Koenraadt CJM, van Oers MM, and Pijlman GP
- Subjects
- Aedes immunology, Animals, Chlorocebus aethiops, DEAD-box RNA Helicases genetics, Gastrointestinal Tract virology, Genome, Viral, Insect Proteins genetics, Salivary Glands virology, Virus Replication, Zika Virus immunology, Zika Virus Infection immunology, Zika Virus Infection virology, Aedes virology, DEAD-box RNA Helicases metabolism, Insect Proteins metabolism, Mosquito Vectors virology, RNA, Viral genetics, Zika Virus genetics, Zika Virus Infection transmission
- Abstract
Zika virus (ZIKV) is an arthropod-borne flavivirus predominantly transmitted by Aedes aegypti mosquitoes and poses a global human health threat. All flaviviruses, including those that exclusively replicate in mosquitoes, produce a highly abundant, noncoding subgenomic flavivirus RNA (sfRNA) in infected cells, which implies an important function of sfRNA during mosquito infection. Currently, the role of sfRNA in flavivirus transmission by mosquitoes is not well understood. Here, we demonstrate that an sfRNA-deficient ZIKV (ZIKVΔSF1) replicates similar to wild-type ZIKV in mosquito cell culture but is severely attenuated in transmission by Ae. aegypti after an infectious blood meal, with 5% saliva-positive mosquitoes for ZIKVΔSF1 vs. 31% for ZIKV. Furthermore, viral titers in the mosquito saliva were lower for ZIKVΔSF1 as compared to ZIKV. Comparison of mosquito infection via infectious blood meals and intrathoracic injections showed that sfRNA is important for ZIKV to overcome the mosquito midgut barrier and to promote virus accumulation in the saliva. Next-generation sequencing of infected mosquitoes showed that viral small-interfering RNAs were elevated upon ZIKVΔSF1 as compared to ZIKV infection. RNA-affinity purification followed by mass spectrometry analysis uncovered that sfRNA specifically interacts with a specific set of Ae. aegypti proteins that are normally associated with RNA turnover and protein translation. The DEAD/H-box helicase ME31B showed the highest affinity for sfRNA and displayed antiviral activity against ZIKV in Ae. aegypti cells. Based on these results, we present a mechanistic model in which sfRNA sequesters ME31B to promote flavivirus replication and virion production to facilitate transmission by mosquitoes., Competing Interests: The authors declare no conflict of interest.
- Published
- 2019
- Full Text
- View/download PDF
49. Herpes Simplex Virus 1 Can Enter Dynamin 1 and 2 Double-Knockout Fibroblasts.
- Author
-
Möckel M, Rahn E, de la Cruz N, Wirtz L, van Lent JWM, Pijlman GP, and Knebel-Mörsdorf D
- Subjects
- Animals, Cells, Cultured, Endocytosis, Gene Knockdown Techniques, Genetic Predisposition to Disease, Host-Pathogen Interactions genetics, Humans, Mice, Semliki forest virus physiology, Dynamin I genetics, Dynamin II genetics, Fibroblasts metabolism, Fibroblasts virology, Herpes Simplex genetics, Herpes Simplex virology, Herpesvirus 1, Human physiology, Virus Internalization
- Abstract
Dynamin GTPases, best known for their role in membrane fission of endocytic vesicles, provide a target for viruses to be exploited during endocytic uptake. Recently, we found that entry of herpes simplex virus 1 (HSV-1) into skin cells depends on dynamin, although our results supported that viral internalization occurs via both direct fusion with the plasma membrane and via endocytic pathways. To further explore the role of dynamin for efficient HSV-1 entry, we utilized conditional dynamin 1 and dynamin 2 double-knockout (DKO) fibroblasts as an experimental tool. Strikingly, HSV-1 entered control and DKO fibroblasts with comparable efficiencies. For comparison, we infected DKO cells with Semliki Forest virus, which is known to adopt clathrin-mediated endocytosis as its internalization pathway, and observed efficient virus entry. These results support the notion that the DKO cells provide alternative pathways for viral uptake. Treatment of cells with the dynamin inhibitor dynasore confirmed that HSV-1 entry depended on dynamin in the control fibroblasts. As expected, dynasore did not interfere with viral entry into DKO cells. Electron microscopy of HSV-1-infected cells suggests viral entry after fusion with the plasma membrane and by endocytosis in both dynamin-expressing and dynamin-deficient cells. Infection at low temperatures where endocytosis is blocked still resulted in HSV-1 entry, although at a reduced level, which suggests that nonendocytic pathways contribute to successful entry. Overall, our results strengthen the impact of dynamin for HSV-1 entry, as only cells that adapt to the lack of dynamin allow dynamin-independent entry. IMPORTANCE The human pathogen herpes simplex virus 1 (HSV-1) can adapt to a variety of cellular pathways to enter cells. In general, HSV-1 is internalized by fusion of its envelope with the plasma membrane or by endocytic pathways, which reflects the high adaptation to differences in its target cells. The challenges are to distinguish whether multiple or only one of these internalization pathways leads to successful entry and, furthermore, to identify the mode of viral uptake. In this study, we focused on dynamin, which promotes endocytic vesicle fission, and explored how the presence and absence of dynamin can influence viral entry. Our results support the idea that HSV-1 entry into mouse embryonic fibroblasts depends on dynamin; however, depletion of dynamin still allows efficient viral entry, suggesting that alternative pathways present upon dynamin depletion can accomplish viral internalization., (Copyright © 2019 American Society for Microbiology.)
- Published
- 2019
- Full Text
- View/download PDF
50. Immunogenicity in Rabbits of Virus-Like Particles from a Contemporary Rabbit Haemorrhagic Disease Virus Type 2 (GI.2/RHDV2/b) Isolated in The Netherlands.
- Author
-
Miao Q, Qi R, Veldkamp L, Ijzer J, Kik ML, Zhu J, Tang A, Dong D, Shi Y, van Oers MM, Liu G, and Pijlman GP
- Subjects
- Animals, Antibodies, Viral blood, Antibody Formation, Baculoviridae, Caliciviridae Infections immunology, Caliciviridae Infections prevention & control, Cluster Analysis, Cytokines analysis, Genetic Vectors, Hemorrhagic Disease Virus, Rabbit classification, Hemorrhagic Disease Virus, Rabbit genetics, Hemorrhagic Disease Virus, Rabbit isolation & purification, Immunity, Cellular, Netherlands, Phylogeny, Rabbits, Sequence Analysis, DNA, Sequence Homology, Sf9 Cells, Spodoptera, Vaccines, Virus-Like Particle administration & dosage, Vaccines, Virus-Like Particle genetics, Virosomes genetics, Caliciviridae Infections veterinary, Hemorrhagic Disease Virus, Rabbit immunology, Vaccines, Virus-Like Particle immunology, Virosomes immunology
- Abstract
Rabbit haemorrhagic disease virus (RHDV) type 2 (GI.2/RHDV2/b) is an emerging pathogen in wild rabbits and in domestic rabbits vaccinated against RHDV (GI.1). Here we report the genome sequence of a contemporary RHDV2 isolate from the Netherlands and investigate the immunogenicity of virus-like particles (VLPs) produced in insect cells. RHDV2 RNA was isolated from the liver of a naturally infected wild rabbit and the complete viral genome sequence was assembled from sequenced RT-PCR products. Phylogenetic analysis based on the VP60 capsid gene demonstrated that the RHDV2 NL2016 isolate clustered with other contemporary RHDV2 strains. The VP60 gene was cloned in a baculovirus expression vector to produce VLPs in Sf9 insect cells. Density-gradient purified RHDV2 VLPs were visualized by transmission electron microscopy as spherical particles of around 30 nm in diameter with a morphology resembling authentic RHDV. Immunization of rabbits with RHDV2 VLPs resulted in high production of serum antibodies against VP60, and the production of cytokines (IFN-γ and IL-4) was significantly elevated in the immunized rabbits compared to the control group. The results demonstrate that the recombinant RHDV2 VLPs are highly immunogenic and may find applications in serological detection assays and might be further developed as a vaccine candidate to protect domestic rabbits against RHDV2 infection., Competing Interests: The authors declare no conflict of interest.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.