20,616 results on '"Virologie"'
Search Results
202. Identification of the potential active site of the septal peptidoglycan polymerase FtsW
- Author
-
Virologie, Sub Membrane Biochemistry & Biophysics, Membrane Biochemistry and Biophysics, Li, Ying, Boes, Adrien, Cui, Yuanyuan, Zhao, Shan, Liao, Qingzhen, Gong, Han, Breukink, Eefjan, Lutkenhaus, Joe, Terrak, Mohammed, Du, Shishen, Virologie, Sub Membrane Biochemistry & Biophysics, Membrane Biochemistry and Biophysics, Li, Ying, Boes, Adrien, Cui, Yuanyuan, Zhao, Shan, Liao, Qingzhen, Gong, Han, Breukink, Eefjan, Lutkenhaus, Joe, Terrak, Mohammed, and Du, Shishen
- Published
- 2021
203. Structural insights into the cross-neutralization of SARS-CoV and SARS-CoV-2 by the human monoclonal antibody 47D11
- Author
-
Sub Structural Biochemistry, Virologie, dI&I I&I-1, Fedry, Juliette, Hurdiss, Daniel L, Wang, Chunyan, Li, Wentao, Obal, Gonzalo, Drulyte, Ieva, Du, Wenjuan, Howes, Stuart C, van Kuppeveld, Frank J M, Förster, Friedrich, Bosch, Berend-Jan, Sub Structural Biochemistry, Virologie, dI&I I&I-1, Fedry, Juliette, Hurdiss, Daniel L, Wang, Chunyan, Li, Wentao, Obal, Gonzalo, Drulyte, Ieva, Du, Wenjuan, Howes, Stuart C, van Kuppeveld, Frank J M, Förster, Friedrich, and Bosch, Berend-Jan
- Published
- 2021
204. Synthetic O-acetylated sialosides facilitate functional receptor identification for human respiratory viruses
- Author
-
Afd Chemical Biology and Drug Discovery, Virologie, dI&I I&I-1, Sub Chemical Biology and Drug Discovery, Chemical Biology and Drug Discovery, Li, Zeshi, Lang, Yifei, Liu, Lin, Bunyatov, Mehman I, Sarmiento, Angelic Isaza, de Groot, Raoul J, Boons, Geert-Jan, Afd Chemical Biology and Drug Discovery, Virologie, dI&I I&I-1, Sub Chemical Biology and Drug Discovery, Chemical Biology and Drug Discovery, Li, Zeshi, Lang, Yifei, Liu, Lin, Bunyatov, Mehman I, Sarmiento, Angelic Isaza, de Groot, Raoul J, and Boons, Geert-Jan
- Published
- 2021
205. Respiratory mucus as a virus-host range determinant
- Author
-
Virologie, dI&I I&I-1, Wallace, Louisa E, Liu, Mengying, van Kuppeveld, Frank, de Vries, Erik, de Haan, C.A.M., Virologie, dI&I I&I-1, Wallace, Louisa E, Liu, Mengying, van Kuppeveld, Frank, de Vries, Erik, and de Haan, C.A.M.
- Published
- 2021
206. SARS-CoV-2 neutralizing human antibodies protect against lower respiratory tract disease in a hamster model
- Author
-
dI&I I&I-1, Virologie, Haagmans, Bart L, Noack, Danny, Okba, Nisreen M A, Li, Wentao, Wang, Chunyan, Bestebroer, Theo, de Vries, Rory, Herfst, Sander, de Meulder, Dennis, Verveer, Elwin, van Run, Peter, Lamers, Mart M, Rijnders, Bart, Rokx, Casper, van Kuppeveld, Frank, Grosveld, Frank, Drabek, Dubravka, GeurtsvanKessel, Corine, Koopmans, Marion, Bosch, Berend Jan, Kuiken, Thijs, Rockx, Barry, dI&I I&I-1, Virologie, Haagmans, Bart L, Noack, Danny, Okba, Nisreen M A, Li, Wentao, Wang, Chunyan, Bestebroer, Theo, de Vries, Rory, Herfst, Sander, de Meulder, Dennis, Verveer, Elwin, van Run, Peter, Lamers, Mart M, Rijnders, Bart, Rokx, Casper, van Kuppeveld, Frank, Grosveld, Frank, Drabek, Dubravka, GeurtsvanKessel, Corine, Koopmans, Marion, Bosch, Berend Jan, Kuiken, Thijs, and Rockx, Barry
- Published
- 2021
207. Structural basis for broad coronavirus neutralization
- Author
-
Virologie, dI&I I&I-1, Sauer, Maximilian M, Tortorici, M Alejandra, Park, Young-Jun, Walls, Alexandra C, Homad, Leah, Acton, Oliver J, Bowen, John E, Wang, Chunyan, Xiong, Xiaoli, de van der Schueren, Willem, Quispe, Joel, Hoffstrom, Benjamin G, Bosch, Berend-Jan, McGuire, Andrew T, Veesler, David, Virologie, dI&I I&I-1, Sauer, Maximilian M, Tortorici, M Alejandra, Park, Young-Jun, Walls, Alexandra C, Homad, Leah, Acton, Oliver J, Bowen, John E, Wang, Chunyan, Xiong, Xiaoli, de van der Schueren, Willem, Quispe, Joel, Hoffstrom, Benjamin G, Bosch, Berend-Jan, McGuire, Andrew T, and Veesler, David
- Published
- 2021
208. Zoonoses Anticipation and Preparedness Initiative, stakeholders conference, February 4 & 5, 2021
- Author
-
Virologie, dI&I I&I-1, LS Infectiebiologie (Bacteriologie), Bedrijfsvoering, Beer, Martin, Amery, Leanne, Bosch, Berend-Jan, Brix, Alexander, Daramola, Olalekan, Inman, Sophie, Jungbäck, Carmen, Kortekaas, Jeroen, Lindo, Viv, Okorji-Obike, Uche, Rodriguez-Conde, Sara, Tang, Alison, Tchelet, Ronen, Vandeputte, Joris, Wichgers Schreur, Paul J, Osterhaus, Ab, Haagmans, Bart, Audonnet, Jean-Christophe, Virologie, dI&I I&I-1, LS Infectiebiologie (Bacteriologie), Bedrijfsvoering, Beer, Martin, Amery, Leanne, Bosch, Berend-Jan, Brix, Alexander, Daramola, Olalekan, Inman, Sophie, Jungbäck, Carmen, Kortekaas, Jeroen, Lindo, Viv, Okorji-Obike, Uche, Rodriguez-Conde, Sara, Tang, Alison, Tchelet, Ronen, Vandeputte, Joris, Wichgers Schreur, Paul J, Osterhaus, Ab, Haagmans, Bart, and Audonnet, Jean-Christophe
- Published
- 2021
209. PKR activity modulation by phosphomimetic mutations of serine residues located three aminoacids upstream of double-stranded RNA binding motifs
- Author
-
UCL - SSS/DDUV/VIRO - Virologie, Cesaro, Teresa, Hayashi, Yohei, Borghese, Fabian, Vertommen, Didier, Wavreil, Fanny, Michiels, Thomas, UCL - SSS/DDUV/VIRO - Virologie, Cesaro, Teresa, Hayashi, Yohei, Borghese, Fabian, Vertommen, Didier, Wavreil, Fanny, and Michiels, Thomas
- Abstract
Eukaryotic translation initiation factor 2 alpha kinase 2 (EIF2AK2), better known as PKR, plays a key role in the response to viral infections and cellular homeostasis by regulating mRNA translation. Upon binding dsRNA, PKR is activated through homodimerization and subsequent autophosphorylation on residues Thr446 and Thr451. In this study, we identified a novel PKR phosphorylation site, Ser6, located 3 amino acids upstream of the first double-stranded RNA binding motif (DRBM1). Another Ser residue occurs in PKR at position 97, the very same position relative to the DRBM2. Ser or Thr residues also occur 3 amino acids upstream DRBMs of other proteins such as ADAR1 or DICER. Phosphoinhibiting mutations (Ser-to-Ala) introduced at Ser6 and Ser97 spontaneously activated PKR. In contrast, phosphomimetic mutations (Ser-to-Asp) inhibited PKR activation following either poly (I:C) transfection or virus infection. These mutations moderately affected dsRNA binding or dimerization, suggesting a model where negative charges occurring at position 6 and 97 tighten the interaction of DRBMs with the kinase domain, thus keeping PKR in an inactive closed conformation even in the presence of dsRNA. This study provides new insights on PKR regulation mechanisms and identifies Ser6 and Ser97 as potential targets to modulate PKR activity for therapeutic purposes.
- Published
- 2021
210. Nucleocytoplasmic Trafficking Perturbation Induced by Picornaviruses
- Author
-
UCL - SSS/DDUV/VIRO - Virologie, Lizcano-Perret, Belén, Michiels, Thomas, UCL - SSS/DDUV/VIRO - Virologie, Lizcano-Perret, Belén, and Michiels, Thomas
- Abstract
Picornaviruses are positive-stranded RNA viruses. Even though replication and translation of their genome take place in the cytoplasm, these viruses evolved different strategies to disturb nucleocytoplasmic trafficking of host proteins and RNA. The major targets of picornavirus are the phenylalanine-glycine (FG)-nucleoporins, which form a mesh in the central channel of the nuclear pore complex through which protein cargos and karyopherins are actively transported in both directions. Interestingly, while enteroviruses use the proteolytic activity of their 2A protein to degrade FG-nucleoporins, cardioviruses act by triggering phosphorylation of these proteins by cellular kinases. By targeting the nuclear pore complex, picornaviruses recruit nuclear proteins to the cytoplasm, where they increase viral genome translation and replication; they affect nuclear translocation of cytoplasmic proteins such as transcription factors that induce innate immune responses and retain host mRNA in the nucleus thereby preventing cell emergency responses and likely making the ribosomal machinery available for translation of viral RNAs.
- Published
- 2021
211. PKR regulation by phosphorylation and antiviral activity of the PKR-ADAR1 axis
- Author
-
UCL - SSS/DDUV/VIRO - Virologie, UCL - Faculté de pharmacie et des sciences biomédicales, Michiels, Thomas, Vikkula, Miikka, Decottignies, Anabelle, Van Baren, Nicolas, Lauwerys, Bernard, Meurs, Eliane, Vanderplasschen, Alain, Cesaro, Teresa, UCL - SSS/DDUV/VIRO - Virologie, UCL - Faculté de pharmacie et des sciences biomédicales, Michiels, Thomas, Vikkula, Miikka, Decottignies, Anabelle, Van Baren, Nicolas, Lauwerys, Bernard, Meurs, Eliane, Vanderplasschen, Alain, and Cesaro, Teresa
- Abstract
Protein kinase RNA-activated (PKR) and Double-stranded RNA-specific adenosine deaminase (ADAR1) are two double stranded RNA binding proteins, respectively involved in the antiviral response to viruses and in the metabolism of dsRNA molecules. PKR is a cellular protein kinase, that in response to dsRNA molecules generated during viral infections, gets activated and phosphorylates the translation initiation factor, eIF2a, leading to translational shutoff and apoptosis. As PKR thereby acts as a potent antiviral effector, many viruses evolved mechanisms to counteract its antiviral response. Previous studies showed that Theiler’s murine encephalomyelitis virus (TMEV), a cardiovirus belonging to the Picornaviridae family, can block PKR activation through the activity of its Leader (L) protein, an accessory protein of the virus known to block IFN gene transcription and perturb nucleocytoplasmic trafficking. In the first part of this thesis I contributed to a work showing that the L protein likely renders PKR insensitive to dsRNA molecules, possibly through the activation of cellular kinases. Next, we analyzed PKR phosphorylation modifications in the hope to identify potential phosphorylation sites responsible for PKR inhibition by TMEV L. We observed that the Ser6 residue located 3aa before the first double-stranded RNA binding motif (DRBM1) of PKR could be phosphorylated. A phospho-mimetic mutation of this site was inhibiting PKR activation after poly(I:C) transfection or viral infection, especially when combined to a phospho-mimetic mutation of the Ser97 residue, located 3aa before the second double- stranded RNA binding motif (DRBM2). We propose a model according to which phosphorylation occurring upstream of DRBMs would tighten the interaction of the DRBMs with the catalytic domain, blocking PKR in a closed conformation, and making it unable to be activated. ADAR1 is an editing enzyme, causing deamination of adenosines into inosines in dsRNA molecules, thus destabilizi, (BIFA - Sciences biomédicales et pharmaceutiques) -- UCL, 2021
- Published
- 2021
212. Second sialic acid-binding site of influenza A virus neuraminidase: Binding receptors for efficient release
- Author
-
Virologie, dI&I I&I-1, Du, Wenjuan, de Vries, Erik, van Kuppeveld, Frank J M, Matrosovich, Mikhail, de Haan, Cornelis A M, Virologie, dI&I I&I-1, Du, Wenjuan, de Vries, Erik, van Kuppeveld, Frank J M, Matrosovich, Mikhail, and de Haan, Cornelis A M
- Published
- 2021
213. Changes in SARS-CoV-2 Spike versus Nucleoprotein Antibody Responses Impact the Estimates of Infections in Population-Based Seroprevalence Studies
- Author
-
Virologie, dI&I I&I-1, Fenwick, Craig, Croxatto, Antony, Coste, Alix T, Pojer, Florence, André, Cyril, Pellaton, Céline, Farina, Alex, Campos, Jérémy, Hacker, David, Lau, Kelvin, Bosch, Berend-Jan, Gonseth Nussle, Semira, Bochud, Murielle, D'Acremont, Valerie, Trono, Didier, Greub, Gilbert, Pantaleo, Giuseppe, Virologie, dI&I I&I-1, Fenwick, Craig, Croxatto, Antony, Coste, Alix T, Pojer, Florence, André, Cyril, Pellaton, Céline, Farina, Alex, Campos, Jérémy, Hacker, David, Lau, Kelvin, Bosch, Berend-Jan, Gonseth Nussle, Semira, Bochud, Murielle, D'Acremont, Valerie, Trono, Didier, Greub, Gilbert, and Pantaleo, Giuseppe
- Published
- 2021
214. Les PU-boxes intragénique contrôle la transcription et la réplication du VIH-1 dans les lignées myéloïdes.
- Author
-
XXIIIèmes Journées Francophones de Virologie (28/29 avril 2021: Montpellier), Hernalsteens, Olivier, Verdikt, Roxane, Vanhulle, Caroline, Delacourt, Nadège, Monceaux, Valerie, Galons, Hervé, Verhasselt, Bruno, Sáez-Cirión, Asier, Van Lint, Carine, XXIIIèmes Journées Francophones de Virologie (28/29 avril 2021: Montpellier), Hernalsteens, Olivier, Verdikt, Roxane, Vanhulle, Caroline, Delacourt, Nadège, Monceaux, Valerie, Galons, Hervé, Verhasselt, Bruno, Sáez-Cirión, Asier, and Van Lint, Carine
- Abstract
info:eu-repo/semantics/published
- Published
- 2021
215. Identification of a new regulator of Bovine Leukemia Virus transcription
- Author
-
XXIIIemes Journées Francophones de Virologie (26-27 avril 2021: Montpellier), Plant, Estelle, Bellefroid, Maxime, Ait-Ammar, Amina, Galais, Mathilde, Rodari, Anthony, Nestola, Lorena, Vanhulle, Caroline, Van Lint, Carine, XXIIIemes Journées Francophones de Virologie (26-27 avril 2021: Montpellier), Plant, Estelle, Bellefroid, Maxime, Ait-Ammar, Amina, Galais, Mathilde, Rodari, Anthony, Nestola, Lorena, Vanhulle, Caroline, and Van Lint, Carine
- Abstract
info:eu-repo/semantics/published
- Published
- 2021
216. Antibacterial resistance, genes encoding toxins and genetic background among Staphylococcus aureus isolated from community-acquired skin and soft tissue infections in France: a national prospective survey
- Author
-
Lamy, B., Laurent, F., Gallon, O., Doucet-Populaire, F., Etienne, J., Decousser, J.-W., and The Collège de Bactériologie Virologie Hygiène (ColBVH) Study Group
- Published
- 2012
- Full Text
- View/download PDF
217. Older adults lack SARS CoV-2 cross-reactive T lymphocytes directed to human coronaviruses OC43 and NL63
- Author
-
Saletti, Giulietta, Gerlach, Thomas, Jansen, Janina M, Molle, Antonia, Elbahesh, Husni, Ludlow, Martin, Li, Wentao, Bosch, Berend-Jan, Osterhaus, Albert D M E, Rimmelzwaan, Guus F, Virologie, dI&I I&I-1, and Bedrijfsvoering
- Subjects
body regions ,viruses ,fungi ,virus diseases ,skin and connective tissue diseases - Abstract
Currently, infections with SARS-Coronavirus-2 (SARS-CoV-2), the causative agent of the COVID-19 pandemic, are responsible for substantial morbidity and mortality worldwide. Older adults subjects > 60 years of age account for > 95% of the over one million fatal cases reported to date. It is unclear why in this age group SARS-CoV-2 infection causes more severe disease than in young adults. We hypothesized that differences in SARS-CoV-2 cross-reactive cellular immunity induced after infection with human coronaviruses (HCoVs), like OC43 and NL63, were at the basis of the differential mortality (and morbidity) observed after SARS-CoV-2 infection, because a small proportion of HCoV-specific T cells cross-react with SARS-CoV-2. Our data demonstrate that pre-existing T cell immunity induced by circulating human alpha- and beta-HCoVs is present in young adult individuals, but virtually absent in older adult subjects. Consequently, the frequency of cross-reactive T cells directed to the novel pandemic SARS-CoV-2 was minimal in most older adults. To the best of our knowledge, this is the first time that the presence of cross-reactive T cells to SARS-CoV-2 is compared in young and older adults. Our findings provide at least a partial explanation for the more severe clinical outcome of SARS-CoV-2 infection observed in the elderly. Moreover, this information could help to design efficacious vaccines for this age group, aiming at the induction of cell-mediated immunity.
- Published
- 2020
218. Structural insights into the cross-neutralization of SARS-CoV and SARS-CoV-2 by the human monoclonal antibody 47D11
- Author
-
Fedry, Juliette, Hurdiss, Daniel L, Wang, Chunyan, Li, Wentao, Obal, Gonzalo, Drulyte, Ieva, Du, Wenjuan, Howes, Stuart C, van Kuppeveld, Frank J M, Förster, Friedrich, Bosch, Berend-Jan, Sub Structural Biochemistry, Virologie, dI&I I&I-1, Sub Structural Biochemistry, Virologie, and dI&I I&I-1
- Subjects
Glycan ,Coronavirus disease 2019 (COVID-19) ,medicine.drug_class ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,Monoclonal antibody ,Antibodies, Viral ,Epitope ,03 medical and health sciences ,Structure-Activity Relationship ,0302 clinical medicine ,Structural Biology ,medicine ,Structure–activity relationship ,Humans ,Health and Medicine ,Binding site ,skin and connective tissue diseases ,General ,Research Articles ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,biology ,Chemistry ,SARS-CoV-2 ,fungi ,Antibodies, Monoclonal ,SciAdv r-articles ,Virology ,Antibodies, Neutralizing ,3. Good health ,respiratory tract diseases ,body regions ,Coronavirus ,Severe acute respiratory syndrome-related coronavirus ,biology.protein ,Antibody ,030217 neurology & neurosurgery ,Research Article - Abstract
Cryo-EM reveals the molecular basis of SARS-CoV-2 cross-neutralization by the monoclonal antibody 47D11., The emergence of SARS-CoV-2 antibody escape mutations highlights the urgent need for broadly neutralizing therapeutics. We previously identified a human monoclonal antibody, 47D11, capable of cross-neutralizing SARS-CoV-2 and SARS-CoV and protecting against the associated respiratory disease in an animal model. Here, we report cryo-EM structures of both trimeric spike ectodomains in complex with the 47D11 Fab. 47D11 binds to the closed receptor-binding domain, distal to the ACE2 binding site. The CDRL3 stabilizes the N343 glycan in an upright conformation, exposing a mutationally constrained hydrophobic pocket, into which the CDRH3 loop inserts two aromatic residues. 47D11 stabilizes a partially open conformation of the SARS-CoV-2 spike, suggesting that it could be used effectively in combination with other antibodies targeting the exposed receptor-binding motif. Together, these results reveal a cross-protective epitope on the SARS-CoV-2 spike and provide a structural roadmap for the development of 47D11 as a prophylactic or postexposure therapy for COVID-19.
- Published
- 2021
219. A reverse genetics system for avian coronavirus infectious bronchitis virus based on targeted RNA recombination
- Author
-
van Beurden, Steven J, Berends, Alinda J, Krämer-Kühl, Annika, Spekreijse, Dieuwertje, Chénard, Gilles, Philipp, Hans-Christian, Mundt, Egbert, Rottier, Peter J M, Verheije, M Hélène, LS Virologie, dI&I I&I-1, dPB I&I, LS Virologie, dI&I I&I-1, and dPB I&I
- Subjects
0301 basic medicine ,Embryonated eggs ,animal structures ,030106 microbiology ,Infectious bronchitis virus ,medicine.disease_cause ,Recombinant virus ,Poultry ,Cell Line ,Vaccine development ,lcsh:Infectious and parasitic diseases ,03 medical and health sciences ,Mice ,Mouse hepatitis virus ,Virology ,medicine ,Animals ,lcsh:RC109-216 ,Gene ,Coronavirus ,Targeted RNA recombination ,Recombination, Genetic ,biology ,Research ,Avian coronavirus ,Embryonated ,RNA ,Avian infectious bronchitis ,biology.organism_classification ,Chicken ,Reverse genetics ,Reverse Genetics ,030104 developmental biology ,Infectious Diseases ,Reverse genetics system ,Gene Targeting ,embryonic structures ,RNA, Viral ,Chickens - Abstract
Background Avian coronavirus infectious bronchitis virus (IBV) is a respiratory pathogen of chickens that causes severe economic losses in the poultry industry worldwide. Major advances in the study of the molecular biology of IBV have resulted from the development of reverse genetics systems for the highly attenuated, cell culture-adapted, IBV strain Beaudette. However, most IBV strains, amongst them virulent field isolates, can only be propagated in embryonated chicken eggs, and not in continuous cell lines. Methods We established a reverse genetics system for the IBV strain H52, based on targeted RNA recombination in a two-step process. First, a genomic and a chimeric synthetic, modified IBV RNA were co-transfected into non-susceptible cells to generate a recombinant chimeric murinized (m) IBV intermediate (mIBV). Herein, the genomic part coding for the spike glycoprotein ectodomain was replaced by that of the coronavirus mouse hepatitis virus (MHV), allowing for the selection and propagation of recombinant mIBV in murine cells. In the second step, mIBV was used as the recipient. To this end a recombination with synthetic RNA comprising the 3′-end of the IBV genome was performed by introducing the complete IBV spike gene, allowing for the rescue and selection of candidate recombinants in embryonated chicken eggs. Results Targeted RNA recombination allowed for the modification of the 3′-end of the IBV genome, encoding all structural and accessory genes. A wild-type recombinant IBV was constructed, containing several synonymous marker mutations. The in ovo growth kinetics and in vivo characteristics of the recombinant virus were similar to those of the parental IBV strain H52. Conclusions Targeted RNA recombination allows for the generation of recombinant IBV strains that are not able to infect and propagate in continuous cell lines. The ability to introduce specific mutations holds promise for the development of rationally designed live-attenuated IBV vaccines and for studies into the biology of IBV in general. Electronic supplementary material The online version of this article (doi:10.1186/s12985-017-0775-8) contains supplementary material, which is available to authorized users.
- Published
- 2017
220. Aminopeptidase N is not required for porcine epidemic diarrhea virus cell entry
- Author
-
Li, Wentao, Luo, Rui, He, Qigai, van Kuppeveld, Frank J M, Rottier, Peter J M, Bosch, Berend-Jan, dI&I I&I-1, LS Virologie, dI&I I&I-1, and LS Virologie
- Subjects
0301 basic medicine ,Cancer Research ,Human coronavirus 229E ,animal structures ,Swine ,Coronacrisis-Taverne ,Gene Expression ,CD13 Antigens ,medicine.disease_cause ,Virus ,Article ,Microbiology ,Aminopeptidase N ,Cell Line ,HeLa ,03 medical and health sciences ,Gene Knockout Techniques ,Dogs ,Virology ,medicine ,Animals ,Humans ,Receptor ,Coronavirus ,chemistry.chemical_classification ,biology ,Virus cell entry ,Porcine epidemic diarrhea virus ,Virus Internalization ,biology.organism_classification ,Intestinal epithelium ,030104 developmental biology ,Infectious Diseases ,chemistry ,Receptors, Virus ,Glycoprotein ,hormones, hormone substitutes, and hormone antagonists - Abstract
Highlights • Overexpression of porcine APN in cells does not confer susceptibility to PEDV. • Knockout APN expression in PEDV-susceptible cells has no effect on PEDV infection. • Results demonstrate that APN is not essential for PEDV cell entry., Porcine epidemic diarrhea virus (PEDV) is an emerging pathogenic coronavirus that causes a significant economic burden to the swine industry. The virus infects the intestinal epithelium and causes villous atrophy, resulting in diarrhea and dehydration. Interaction of the viral spike (S) surface glycoprotein − through its S1 subunit − with the host cell receptor is the first step in infection and the main determinant for virus tropism. As for several other alphacoronaviruses including the porcine transmissible gastroenteritis virus (TGEV) and the human coronavirus 229E (HCoV-229E), the aminopeptidase N (APN) protein was reported to be a functional receptor for PEDV. In this study we examined the role of APN as a receptor. We show that overexpression of porcine APN renders MDCK cells susceptible to TGEV, but not to PEDV. Consistently, unlike TGEV-S1, PEDV-S1 exhibited no binding to cell-surface expressed APN or to a soluble version of APN. Moreover, preincubation of these viruses with soluble APN or pretreatment of APN expressing ST cells with soluble TGEV-S1 blocked TGEV infection, but had no effect on infection by PEDV. The combined observations indicated that APN is not required for PEDV infection. To definitively prove this conclusion, we applied CRISPR/Cas9 genome engineering to knock out APN expression in PEDV-susceptible porcine (ST) and human cell lines (Huh7 and HeLa). As a consequence these cells no longer bound TGEV-S1 and HCoV-229E-S1 at their surface and were resistant to infection by the corresponding viruses. However, genetic ablation of APN expression had no effect on their infectability by PEDV, demonstrating that APN is not essential for PEDV cell entry.
- Published
- 2017
221. In vitro activity of daptomycin against Staphylococci isolated from bacteremia and community-onset skin and soft tissue infections in France: data from two nationwide studies
- Author
-
Gallon, O., Guillet-Caruba, C., Lamy, B., Laurent, F., Doucet-Populaire, F., Decousser, J.-W., and Collège de Bactériologie Virologie Hygiène Study Group (ColBVH)
- Published
- 2009
- Full Text
- View/download PDF
222. Development and Validation of a S1 Protein-Based ELISA for the Specific Detection of Antibodies against Equine Coronavirus
- Author
-
Zhao, Shan, Smits, Constance, Schuurman, Nancy, Barnum, Samantha, Pusterla, Nicola, Kuppeveld, Frank van, Bosch, Berend-Jan, Maanen, Kees van, Egberink, Herman, LS Virologie, dI&I I&I-1, LS Klinisch Onderzoek Wagenaar, dI&I I&I-4, LS Virologie, dI&I I&I-1, LS Klinisch Onderzoek Wagenaar, and dI&I I&I-4
- Subjects
0301 basic medicine ,Iceland ,Seroprevalence ,Antibodies, Viral ,Equine coronavirus ,Spike S1 protein ,0403 veterinary science ,Seroepidemiologic Studies ,Medicine ,Betacoronavirus 1 ,Viral ,Neutralizing ,Netherlands ,biology ,seroprevalence ,equine coronavirus ,04 agricultural and veterinary sciences ,Virus neutralization ,Spike Glycoprotein ,Infectious Diseases ,Spike Glycoprotein, Coronavirus ,virus neutralization ,ELISA ,Antibody ,Coronavirus Infections ,Infection ,040301 veterinary sciences ,Specific detection ,Virus Neutralization ,Enzyme-Linked Immunosorbent Assay ,Microbiology ,Article ,Antibodies ,Vaccine Related ,03 medical and health sciences ,Virology ,Biodefense ,Animals ,Serologic Tests ,Horses ,Seroconversion ,business.industry ,Prevention ,Outbreak ,Horse ,Antibodies, Neutralizing ,Coronavirus ,spike S1 ,030104 developmental biology ,Emerging Infectious Diseases ,ROC Curve ,biology.protein ,Horse Diseases ,business - Abstract
Equine coronavirus (ECoV) is considered to be involved in enteric diseases in foals. Recently, several outbreaks of ECoV infection have also been reported in adult horses from the USA, France and Japan. Epidemiological studies of ECoV infection are still limited, and the seroprevalence of ECoV infection in Europe is unknown. In this study, an indirect enzyme-linked immunosorbent assay (ELISA) method utilizing ECoV spike S1 protein was developed in two formats, and further validated by analyzing 27 paired serum samples (acute and convalescent sera) from horses involved in an ECoV outbreak and 1084 sera of horses with unknown ECoV exposure. Both formats showed high diagnostic accuracy compared to virus neutralization (VN) assay. Receiver-operating characteristic (ROC) analyses were performed to determine the best cut-off values for both ELISA formats, assuming a test specificity of 99%. Employing the developed ELISA method, we detected seroconversion in 70.4% of horses from an ECoV outbreak. Among the 1084 horse sera, seropositivity varied from 25.9% (young horses) to 82.8% (adult horses) in Dutch horse populations. Further, sera of Icelandic horses were included in this study and a significant number of sera (62%) were found to be positive. Overall, the results demonstrated that the ECoV S1-based ELISA has reliable diagnostic performance compared to the VN assay and is a useful assay to support seroconversion in horses involved with ECoV outbreaks and to estimate ECoV seroprevalence in populations of horses.
- Published
- 2019
223. Fluoxetine Inhibits Enterovirus Replication by Targeting the Viral 2C Protein in a Stereospecific Manner
- Author
-
Bauer, L., Manganaro, Roberto, Zonsics, Birgit, Strating, J.R.P.M., El Kazzi, Priscila, Lorenzo Lopez, Moira, Ulferts, R., van Hoey, Clara, Mate, Maria J., Langer, Thierry, Coutard, Bruno, Brancale, Andrea, van Kuppeveld, F.J.M., LS Virologie, dI&I I&I-1, Department of Infectious Diseases and Immunology [Utrecht, The Netherlands], Utrecht University [Utrecht]-WHO Collaborating Center for Campylobacter /OIE Reference Laboratory for Campylobacteriosis (Utrecht), School of Pharmacy and Pharmaceutical Sciences [Cardiff, UK], Cardiff University, Architecture et fonction des Macromolécules Biologiques - UMR 6098 (AFMB), Université de Provence - Aix-Marseille 1-Centre National de la Recherche Scientifique (CNRS), Department of Pharmaceutical Chemistry [Vienna, Austria], University of Vienna [Vienna], Unité des Virus Emergents (UVE), Institut de Recherche pour le Développement (IRD)-Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Aix Marseille Université (AMU)-Institut de Recherche pour le Développement (IRD)-Institut National de la Santé et de la Recherche Médicale (INSERM), coutard, bruno, LS Virologie, and dI&I I&I-1
- Subjects
Models, Molecular ,0301 basic medicine ,Drug ,[SDV.SP.MED] Life Sciences [q-bio]/Pharmaceutical sciences/Medication ,Protein Conformation ,Viral protein ,media_common.quotation_subject ,viruses ,030106 microbiology ,Viral Nonstructural Proteins ,medicine.disease_cause ,Article ,enteroviruses ,law.invention ,03 medical and health sciences ,[SDV.SP.MED]Life Sciences [q-bio]/Pharmaceutical sciences/Medication ,law ,Fluoxetine ,medicine ,Humans ,Homology modeling ,virus replication ,media_common ,Enterovirus D, Human ,[SDV.MP.VIR] Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,Molecular Structure ,drug repurposing ,Chemistry ,molecular modeling ,Drug Repositioning ,virus diseases ,Virology ,antiviral ,Enterovirus B, Human ,3. Good health ,Drug repositioning ,030104 developmental biology ,Infectious Diseases ,Viral replication ,Structural Homology, Protein ,Mutation ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,Recombinant DNA ,Enterovirus ,Carrier Proteins ,HeLa Cells ,Protein Binding ,medicine.drug - Abstract
International audience; Enteroviruses (family Picornaviridae) comprise a large group of human pathogens against which no licensed antiviral therapy exists. Drug-repurposing screens uncovered the FDA-approved drug fluoxetine as a replication inhibitor of enterovirus B and D species. Fluoxetine likely targets the nonstructural viral protein 2C, but detailed mode-of-action studies are missing because structural information on 2C of fluoxetine-sensitive enteroviruses is lacking. We here show that broad-spectrum anti-enteroviral activity of fluoxetine is stereospecific concomitant with binding to recombinant 2C. (S)-Fluoxetine inhibits with a 5-fold lower 50% effective concentration (EC50) than racemic fluoxetine. Using a homology model of 2C of the fluoxetine-sensitive enterovirus coxsackievirus B3 (CVB3) based upon a recently elucidated structure of a fluoxetine-insensitive enterovirus, we predicted stable binding of (S)-fluoxetine. Structure-guided mutations disrupted binding and rendered coxsackievirus B3 (CVB3) resistant to fluoxetine. The study provides new insights into the anti-enteroviral mode-of-action of fluoxetine. Importantly, using only (S)-fluoxetine would allow for lower dosing in patients, thereby likely reducing side effects.
- Published
- 2019
224. Species-Specific Colocalization of Middle East Respiratory Syndrome Coronavirus Attachment and Entry Receptors
- Author
-
Widagdo, W, Okba, Nisreen M A, Li, Wentao, de Jong, Alwin, de Swart, Rik L, Begeman, Lineke, van den Brand, Judith M A, Bosch, Berend-Jan, Haagmans, Bart L, LS Virologie, dI&I I&I-1, Veterinair Pathologisch Diagnostisch Cnt, dPB I&I, dPB CR, LS Virologie, dI&I I&I-1, Veterinair Pathologisch Diagnostisch Cnt, dPB I&I, dPB CR, and Virology
- Subjects
Camelus ,Swine ,Middle East respiratory syndrome coronavirus ,Alveolar Epithelium ,viruses ,Immunology ,S1A domain ,Coronacrisis-Taverne ,medicine.disease_cause ,Microbiology ,Host Specificity ,Virus ,common pipistrelle bats ,Cell Line ,03 medical and health sciences ,Chiroptera ,Virology ,medicine ,Animals ,Protein Interaction Domains and Motifs ,Receptor ,humans ,Tropism ,030304 developmental biology ,0303 health sciences ,Mucous Membrane ,biology ,030306 microbiology ,Colocalization ,Epithelial Cells ,dromedary camels ,Virus Internalization ,Intestinal epithelium ,Insect Science ,Spike Glycoprotein, Coronavirus ,biology.protein ,Receptors, Virus ,Pathogenesis and Immunity ,Rabbits ,Coronavirus Infections ,Neuraminidase ,Protein Binding - Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) uses the S1(B) domain of its spike protein to bind to dipeptidyl peptidase 4 (DPP4), its functional receptor, and its S1(A) domain to bind to sialic acids. The tissue localization of DPP4 in humans, bats, camelids, pigs, and rabbits generally correlates with MERS-CoV tropism, highlighting the role of DPP4 in virus pathogenesis and transmission. However, MERS-CoV S1(A) does not indiscriminately bind to all α2,3-sialic acids, and the species-specific binding and tissue distribution of these sialic acids in different MERS-CoV-susceptible species have not been investigated. We established a novel method to detect these sialic acids on tissue sections of various organs of different susceptible species by using nanoparticles displaying multivalent MERS-CoV S1(A). We found that the nanoparticles specifically bound to the nasal epithelial cells of dromedary camels, type II pneumocytes in human lungs, and the intestinal epithelial cells of common pipistrelle bats. Desialylation by neuraminidase abolished nanoparticle binding and significantly reduced MERS-CoV infection in primary susceptible cells. In contrast, S1(A) nanoparticles did not bind to the intestinal epithelium of serotine bats and frugivorous bat species, nor did they bind to the nasal epithelium of pigs and rabbits. Both pigs and rabbits have been shown to shed less infectious virus than dromedary camels and do not transmit the virus via either contact or airborne routes. Our results depict species-specific colocalization of MERS-CoV entry and attachment receptors, which may be relevant in the transmission and pathogenesis of MERS-CoV. IMPORTANCE MERS-CoV uses the S1(B) domain of its spike protein to attach to its host receptor, dipeptidyl peptidase 4 (DPP4). The tissue localization of DPP4 has been mapped in different susceptible species. On the other hand, the S1(A) domain, the N-terminal domain of this spike protein, preferentially binds to several glycotopes of α2,3-sialic acids, the attachment factor of MERS-CoV. Here we show, using a novel method, that the S1(A) domain specifically binds to the nasal epithelium of dromedary camels, alveolar epithelium of humans, and intestinal epithelium of common pipistrelle bats. In contrast, it does not bind to the nasal epithelium of pigs or rabbits, nor does it bind to the intestinal epithelium of serotine bats and frugivorous bat species. This finding supports the importance of the S1(A) domain in MERS-CoV infection and tropism, suggests its role in transmission, and highlights its potential use as a component of novel vaccine candidates.
- Published
- 2019
225. Lipid Droplets Grease Enterovirus Replication
- Author
-
Belov, George A, van Kuppeveld, Frank J M, dI&I I&I-1, LS Virologie, dI&I I&I-1, and LS Virologie
- Subjects
viruses ,Cell ,Coronacrisis-Taverne ,macromolecular substances ,Biology ,medicine.disease_cause ,Virus Replication ,Microbiology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Virology ,Lipid droplet ,Organelle ,medicine ,Enterovirus Infections ,Humans ,030304 developmental biology ,Enterovirus ,0303 health sciences ,RNA ,Lipid metabolism ,Replication (microscopy) ,Lipid Droplets ,Lipids ,Cell biology ,medicine.anatomical_structure ,Viral replication ,Parasitology ,030217 neurology & neurosurgery - Abstract
Positive-stranded RNA viruses extensively remodel host cell architecture to enable viral replication. Here we examined the poorly understood formation of specialized membrane compartments which are critical sites for the synthesis of the viral genome. We show that the replication compartments (RCs) of enteroviruses are created through novel membrane contact sites that recruit host lipid droplets (LDs) to the RCs. Viral proteins tether the RCs to the LDs and interact with the host lipolysis machinery to enable transfer of fatty acids from LDs, thereby providing lipids essential for RC biogenesis. Inhibiting the formation of the membrane contact sites between LDs and RCs or inhibition of the lipolysis pathway disrupts RC biogenesis and enterovirus replication. Our data illuminate mechanistic and functional aspects of organelle remodeling in viral infection and establish that pharmacological targeting of contact sites linking viral and host compartments is a potential strategy for antiviral development.
- Published
- 2019
226. Serological Screening for Coronavirus Infections in Cats
- Author
-
Zhao, Shan, Li, Wentao, Schuurman, Nancy, van Kuppeveld, Frank, Bosch, Berend-Jan, Egberink, Herman, LS Virologie, dI&I I&I-1, dI&I I&I-4, LS Klinisch Onderzoek Wagenaar, LS Virologie, dI&I I&I-1, dI&I I&I-4, and LS Klinisch Onderzoek Wagenaar
- Subjects
0301 basic medicine ,Human coronavirus 229E ,viruses ,coronaviruses ,030106 microbiology ,lcsh:QR1-502 ,Cross-species transmission ,Cross Reactions ,Antibodies, Viral ,Cat Diseases ,medicine.disease_cause ,spike protein ,Article ,Epitope ,lcsh:Microbiology ,Serology ,03 medical and health sciences ,Virology ,medicine ,Animals ,Coronavirus, Feline ,cross-reaction ,Coronavirus ,CATS ,biology ,cats ,virus diseases ,cross-species transmission ,biology.organism_classification ,030104 developmental biology ,Infectious Diseases ,Spike Glycoprotein, Coronavirus ,virus neutralization ,biology.protein ,ELISA ,Antibody ,Coronavirus Infections ,Porcine epidemic diarrhea virus - Abstract
Coronaviruses (CoVs) are widespread among mammals and birds and known for their potential for cross-species transmission. In cats, infections with feline coronaviruses (FCoVs) are common. Several non-feline coronaviruses have been reported to infect feline cells as well as cats after experimental infection, supported by their ability to engage the feline receptor ortholog for cell entry. However, whether cats might become naturally infected with CoVs of other species is unknown. We analyzed coronavirus infections in cats by serological monitoring. In total 137 cat serum samples and 25 FCoV type 1 or type 2-specific antisera were screened for the presence of antibodies against the S1 receptor binding subunit of the CoV spike protein, which is immunogenic and possesses low amino acid sequence identity among coronavirus species. Seventy-eight sera were positive for antibodies that recognized one or more coronavirus S1s whereas 1 serum exclusively reacted with human coronavirus 229E (HCoV-229E) and two sera exclusively reacted with porcine delta coronavirus (PDCoV). We observed antigenic cross-reactivity between S1s of type 1 and type 2 FCoVs, and between FCoV type 1 and porcine epidemic diarrhea virus (PEDV). Domain mapping of antibody epitopes indicated the presence of conserved epitope(s) particularly in the CD domains of S1. The cross-reactivity of FCoV type 1 and PEDV was also observed at the level of virus neutralization. To conclude, we provide the first evidence of antigenic cross-reactivity among S1 proteins of coronaviruses, which should be considered in the development of serological diagnoses. In addition, the potential role of cats in cross-species transmission of coronaviruses cannot be excluded.
- Published
- 2019
227. Convergent evolution in the mechanisms of ACBD3 recruitment to picornavirus replication sites
- Author
-
Horova, Vladimira, Lyoo, Heyrhyoung, Różycki, Bartosz, Chalupska, Dominika, Smola, Miroslav, Humpolickova, Jana, Strating, Jeroen R. P. M., van Kuppeveld, Frank J. M., Boura, Evzen, Klima, Martin, LS Virologie, dI&I I&I-1, LS Virologie, and dI&I I&I-1
- Subjects
Models, Molecular ,RNA viruses ,Viral Diseases ,Picornavirus ,Protein Conformation ,viruses ,Sequence Homology ,Picornaviridae ,Crystallography, X-Ray ,Virus Replication ,Pathology and Laboratory Medicine ,medicine.disease_cause ,Physical Chemistry ,Biochemistry ,Enteroviruses ,Medicine and Health Sciences ,Biology (General) ,0303 health sciences ,Crystallography ,biology ,Effector ,Physics ,Poliovirus ,030302 biochemistry & molecular biology ,virus diseases ,Condensed Matter Physics ,3. Good health ,Phosphotransferases (Alcohol Group Acceptor) ,Chemistry ,Infectious Diseases ,Medical Microbiology ,Viral Pathogens ,Host-Pathogen Interactions ,Physical Sciences ,Viruses ,Crystal Structure ,Pathogens ,Rhinovirus ,Crystallization ,Dimerization ,Protein Binding ,Research Article ,Viral protein ,QH301-705.5 ,Immunology ,Microbiology ,Viral Proteins ,03 medical and health sciences ,Protein Domains ,Virology ,Genetics ,medicine ,Humans ,Solid State Physics ,ddc:610 ,Amino Acid Sequence ,Protein Interactions ,Microbial Pathogens ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,Point mutation ,Organisms ,Membrane Proteins ,Biology and Life Sciences ,Proteins ,RC581-607 ,biology.organism_classification ,Viral Replication ,HEK293 Cells ,Enterovirus Infection ,Chemical Properties ,Viral replication ,Mutation ,Enterovirus ,Parasitology ,Immunologic diseases. Allergy - Abstract
Enteroviruses, members of the family of picornaviruses, are the most common viral infectious agents in humans causing a broad spectrum of diseases ranging from mild respiratory illnesses to life-threatening infections. To efficiently replicate within the host cell, enteroviruses hijack several host factors, such as ACBD3. ACBD3 facilitates replication of various enterovirus species, however, structural determinants of ACBD3 recruitment to the viral replication sites are poorly understood. Here, we present a structural characterization of the interaction between ACBD3 and the non-structural 3A proteins of four representative enteroviruses (poliovirus, enterovirus A71, enterovirus D68, and rhinovirus B14). In addition, we describe the details of the 3A-3A interaction causing the assembly of the ACBD3-3A heterotetramers and the interaction between the ACBD3-3A complex and the lipid bilayer. Using structure-guided identification of the point mutations disrupting these interactions, we demonstrate their roles in the intracellular localization of these proteins, recruitment of downstream effectors of ACBD3, and facilitation of enterovirus replication. These structures uncovered a striking convergence in the mechanisms of how enteroviruses and kobuviruses, members of a distinct group of picornaviruses that also rely on ACBD3, recruit ACBD3 and its downstream effectors to the sites of viral replication., Author summary Enteroviruses are the most common viruses infecting humans. They cause a broad spectrum of diseases ranging from common cold to life-threatening diseases, such as poliomyelitis. To date, no effective antiviral therapy for enteroviruses has been approved yet. To ensure efficient replication, enteroviruses hijack several host factors, recruit them to the sites of virus replication, and use their physiological functions for their own purposes. Here, we characterize the complexes composed of the host protein ACBD3 and the ACBD3-binding viral proteins (called 3A) of four representative enteroviruses. Our study reveals the atomic details of these complexes and identifies the amino acid residues important for the interaction. We found out that the 3A proteins of enteroviruses bind to the same regions of ACBD3 as the 3A proteins of kobuviruses, a distinct group of viruses that also rely on ACBD3, but are oriented in the opposite directions. This observation reveals a striking case of convergent evolutionary pathways that have evolved to allow enteroviruses and kobuviruses (which are two distinct groups of the Picornaviridae family) to recruit a common host target, ACBD3, and its downstream effectors to the sites of viral replication.
- Published
- 2019
228. Structural Studies of Coronavirus Fusion Proteins
- Author
-
Walls, Alexandra C., Tortorici, M. Alejandra, Xiong, Xiaoli, Snijder, Joost, Frenz, Brandon, Bosch, Berend-Jan, DiMaio, Frank, Corti, Davide, Rey, Félix A., Veesler, David, Sub Biomol.Mass Spectrometry & Proteom., dI&I I&I-1, LS Virologie, Sub Biomol.Mass Spectrometry & Proteom., dI&I I&I-1, and LS Virologie
- Subjects
Chemistry ,medicine ,Coronacrisis-Taverne ,medicine.disease_cause ,Instrumentation ,Fusion protein ,Virology ,Coronavirus - Published
- 2019
229. Bypassing pan-enterovirus host factor PLA2G16
- Author
-
Baggen, Jim, Liu, Yue, Lyoo, Heyrhyoung, van Vliet, Arno L W, Wahedi, Maryam, de Bruin, Jost W, Roberts, Richard W, Overduin, Pieter, Meijer, Adam, Rossmann, Michael G, Thibaut, Hendrik Jan, van Kuppeveld, Frank J M, LS Virologie, dI&I I&I-1, LS Virologie, and dI&I I&I-1
- Subjects
viruses ,General Physics and Astronomy ,CRYO-EM ,medicine.disease_cause ,Genome ,chemistry.chemical_compound ,Virus Uncoating ,DEFOCUS ,lcsh:Science ,Glycosaminoglycans ,Host factor ,Enterovirus D, Human ,0303 health sciences ,Multidisciplinary ,REFINEMENT ,biology ,MICROSCOPY ,Virus structures ,PHENIX ,3. Good health ,Cell biology ,Multidisciplinary Sciences ,Phospholipases A2, Calcium-Independent ,ENTRY ,Receptors, Virus ,Science & Technology - Other Topics ,Glycan ,Science ,Enterovirus D ,Genome, Viral ,Virus-host interactions ,ANISOTROPIC MAGNIFICATION DISTORTION ,Article ,General Biochemistry, Genetics and Molecular Biology ,Virus ,SCREENS ,03 medical and health sciences ,Cell Line, Tumor ,Enterovirus Infections ,medicine ,Humans ,030304 developmental biology ,Science & Technology ,RECEPTOR ,030306 microbiology ,Tumor Suppressor Proteins ,Cryoelectron Microscopy ,HEK 293 cells ,General Chemistry ,Virus Internalization ,N-Acetylneuraminic Acid ,Sialic acid ,HEK293 Cells ,chemistry ,biology.protein ,Enterovirus ,lcsh:Q ,SYSTEM ,HeLa Cells - Abstract
Enteroviruses are a major cause of human disease. Adipose-specific phospholipase A2 (PLA2G16) was recently identified as a pan-enterovirus host factor and potential drug target. In this study, we identify a possible mechanism of PLA2G16 evasion by employing a dual glycan receptor-binding enterovirus D68 (EV-D68) strain. We previously showed that this strain does not strictly require the canonical EV-D68 receptor sialic acid. Here, we employ a haploid screen to identify sulfated glycosaminoglycans (sGAGs) as its second glycan receptor. Remarkably, engagement of sGAGs enables this virus to bypass PLA2G16. Using cryo-EM analysis, we reveal that, in contrast to sialic acid, sGAGs stimulate genome release from virions via structural changes that enlarge the putative openings for genome egress. Together, we describe an enterovirus that can bypass PLA2G16 and identify additional virion destabilization as a potential mechanism to circumvent PLA2G16., The adipose-specific phospholipase A2 (PLA2G16) has been identified as a pan-enterovirus host factor. Here, the authors show that the enterovirus EV-D68-947 can bypass PLA2G16 by using sulfated glycosaminoglycans as alternative glycan receptors and that the interaction induces structural changes promoting viral genome release.
- Published
- 2019
230. Enhanced Inhibition of Influenza A Virus Adhesion by Di- and Trivalent Hemagglutinin Inhibitors
- Author
-
Lu, Wenjing, Du, Wenjuan, Somovilla, Victor J, Yu, Guangyun, Haksar, Diksha, de Vries, Erik, Boons, Geert-Jan, de Vries, Robert P, de Haan, Cornelis A M, Pieters, Roland J, Afd Chemical Biology and Drug Discovery, dI&I I&I-1, LS Virologie, Sub Chemical Biology and Drug Discovery, Chemical Biology and Drug Discovery, Afd Chemical Biology and Drug Discovery, dI&I I&I-1, LS Virologie, Sub Chemical Biology and Drug Discovery, and Chemical Biology and Drug Discovery
- Subjects
Hemagglutinin (influenza) ,Hemagglutinin Glycoproteins, Influenza Virus ,Ligands ,medicine.disease_cause ,Antiviral Agents ,01 natural sciences ,Adhesion protein ,Madin Darby Canine Kidney Cells ,03 medical and health sciences ,Dogs ,Influenza A Virus, H1N1 Subtype ,Drug Discovery ,Influenza A virus ,medicine ,Animals ,030304 developmental biology ,0303 health sciences ,biology ,Chemistry ,Madin Darby canine kidney cell ,Adhesion ,Carbohydrate ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Carbohydrate Sequence ,Biochemistry ,Sialic Acids ,biology.protein ,Molecular Medicine ,Glycoconjugates - Abstract
[Image: see text] Multivalent carbohydrate-based ligands were synthesized and evaluated as inhibitors of the adhesion protein HA of the influenza A virus (IAV). HA relies on multivalency for strong viral adhesion. While viral adhesion inhibition by large polymeric molecules has proven viable, limited success was reached for smaller multivalent compounds. By linking of sialylated LAcNAc units to di- and trivalent scaffolds, inhibitors were obtained with an up to 428-fold enhanced inhibition in various assays.
- Published
- 2019
231. Identification of the Cell-Surface Protease ADAM9 as an Entry Factor for Encephalomyocarditis Virus
- Author
-
Baggen, Jim, Thibaut, Hendrik Jan, Hurdiss, Daniel L, Wahedi, Maryam, Marceau, Caleb D, van Vliet, Arno L W, Carette, Jan E, van Kuppeveld, Frank J M, LS Virologie, dI&I I&I-1, LS Virologie, and dI&I I&I-1
- Subjects
Picornavirus ,viruses ,ved/biology.organism_classification_rank.species ,disintegrin and metalloproteinase domain-containing protein 9 (adam9) ,Virus Attachment ,Virus Replication ,Microbiology ,Virus ,Host-Microbe Biology ,Gene Knockout Techniques ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Virology ,INFECTION ,Cardiovirus Infections ,Animals ,Humans ,PIGS ,Model organism ,Receptor ,Pathogen ,haploid genetic screen ,030304 developmental biology ,REQUIRES ,0303 health sciences ,METALLOPROTEASE-DISINTEGRIN MDC9 ,Science & Technology ,biology ,RECEPTOR ,Genome, Human ,ved/biology ,Fibroblast growth factor receptor 1 ,Cell Membrane ,Membrane Proteins ,Virus Internalization ,biology.organism_classification ,encephalomyocarditis virus ,QR1-502 ,3. Good health ,ADAM Proteins ,disintegrin and metalloproteinase domain-containing protein 9 (ADAM9) ,030220 oncology & carcinogenesis ,Signal transduction ,Life Sciences & Biomedicine ,Research Article ,Genetic screen - Abstract
EMCV is an animal pathogen that causes acute viral infections, usually myocarditis or encephalitis. It is thought to circulate mainly among rodents, from which it is occasionally transmitted to other animal species, including humans. EMCV causes fatal outbreaks of myocarditis and encephalitis in pig farms and zoos, making it an important veterinary pathogen. Although EMCV has been widely used as a model to study mechanisms of viral disease in mice, little is known about its entry mechanism. Here, we employ a haploid genetic screen for EMCV host factors and identify an essential role for ADAM9 in EMCV entry., Encephalomyocarditis virus (EMCV) is an animal pathogen and an important model organism, whose receptor requirements are poorly understood. Here, we employed a genome-wide haploid genetic screen to identify novel EMCV host factors. In addition to the previously described picornavirus receptors sialic acid and glycosaminoglycans, this screen unveiled important new host factors for EMCV. These factors include components of the fibroblast growth factor (FGF) signaling pathway, such as the potential receptors FGFR1 and ADAM9, a cell-surface metalloproteinase. By employing various knockout cells, we confirmed the importance of the identified host factors for EMCV infection. The largest reduction in infection efficiency was observed in cells lacking ADAM9. Pharmacological inhibition of the metalloproteinase activity of ADAM9 did not affect virus infection. Moreover, reconstitution of inactive ADAM9 in knockout cells restored susceptibility to EMCV, pointing to a proteinase-independent role of ADAM9 in mediating EMCV infection. Using neutralization assays with ADAM9-specific antiserum and soluble receptor proteins, we provided evidence for a role of ADAM9 in EMCV entry. Finally, binding assays showed that ADAM9 facilitates attachment of EMCV to the cell surface. Together, our findings reveal a role for ADAM9 as a novel receptor or cofactor for EMCV.
- Published
- 2019
232. Host factor prioritization for pan-viral genetic perturbation screens using random intercept models and network propagation
- Author
-
Dirmeier, Simon, Dächert, Christopher, van Hemert, Martijn, Tas, Ali, Ogando, Natacha S, van Kuppeveld, Frank, Bartenschlager, Ralf, Kaderali, Lars, Binder, Marco, Beerenwinkel, Niko, Virologie, dI&I I&I-1, Virologie, and dI&I I&I-1
- Subjects
0301 basic medicine ,RNA viruses ,Genetic Screens ,Genes, Viral ,Gene regulatory network ,Gene Identification and Analysis ,Hepacivirus ,Genetic Networks ,Dengue virus ,medicine.disease_cause ,Pathology and Laboratory Medicine ,Biochemistry ,0302 clinical medicine ,RNA interference ,Viral replication ,Medicine and Health Sciences ,Gene Regulatory Networks ,Small interfering RNAs ,Biology (General) ,Host factor ,Ecology ,Hepatitis C virus ,Genetic screens ,Nucleic acids ,Genetic networks ,Computational Theory and Mathematics ,Genetic interference ,Medical Microbiology ,Modeling and Simulation ,Viral Pathogens ,Viruses ,Epigenetics ,Pathogens ,Network Analysis ,Research Article ,Computer and Information Sciences ,QH301-705.5 ,Computational biology ,Biology ,Models, Biological ,Microbiology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Interaction network ,ssRNA viruses ,Small interfering RNA ,Modelling and Simulation ,Virology ,medicine ,Genetics ,Non-coding RNA ,Gene ,Molecular Biology ,Microbial Pathogens ,Ecology, Evolution, Behavior and Systematics ,Host Microbial Interactions ,Flaviviruses ,Organisms ,Biology and Life Sciences ,Dengue Virus ,Viral Replication ,Hepatitis viruses ,Gene regulation ,030104 developmental biology ,RNA ,Gene expression ,030217 neurology & neurosurgery ,Genetic screen - Abstract
Genetic perturbation screens using RNA interference (RNAi) have been conducted successfully to identify host factors that are essential for the life cycle of bacteria or viruses. So far, most published studies identified host factors primarily for single pathogens. Furthermore, often only a small subset of genes, e.g., genes encoding kinases, have been targeted. Identification of host factors on a pan-pathogen level, i.e., genes that are crucial for the replication of a diverse group of pathogens has received relatively little attention, despite the fact that such common host factors would be highly relevant, for instance, for devising broad-spectrum anti-pathogenic drugs. Here, we present a novel two-stage procedure for the identification of host factors involved in the replication of different viruses using a combination of random effects models and Markov random walks on a functional interaction network. We first infer candidate genes by jointly analyzing multiple perturbations screens while at the same time adjusting for high variance inherent in these screens. Subsequently the inferred estimates are spread across a network of functional interactions thereby allowing for the analysis of missing genes in the biological studies, smoothing the effect sizes of previously found host factors, and considering a priori pathway information defined over edges of the network. We applied the procedure to RNAi screening data of four different positive-sense single-stranded RNA viruses, Hepatitis C virus, Chikungunya virus, Dengue virus and Severe acute respiratory syndrome coronavirus, and detected novel host factors, including UBC, PLCG1, and DYRK1B, which are predicted to significantly impact the replication cycles of these viruses. We validated the detected host factors experimentally using pharmacological inhibition and an additional siRNA screen and found that some of the predicted host factors indeed influence the replication of these pathogens, PLoS Computational Biology, 16 (2), ISSN:1553-734X, ISSN:1553-7358
- Published
- 2019
233. Origins of Enterovirus Replication Organelles Established by Whole-Cell Electron Microscopy
- Author
-
Melia, Charlotte E, Peddie, Christopher J, de Jong, Anja W M, Snijder, Eric J, Collinson, Lucy M, Koster, Abraham J, van der Schaar, Hilde M, van Kuppeveld, Frank J M, Bárcena, Montserrat, LS Virologie, dI&I I&I-1, LS Virologie, and dI&I I&I-1
- Subjects
Picornavirus ,Image Processing ,Golgi Apparatus ,Endoplasmic Reticulum/ultrastructure ,Enterovirus/physiology ,Endoplasmic Reticulum ,Virus Replication ,Computer-Assisted ,Lipid droplet ,volume electron microscopy ,Chlorocebus aethiops ,Image Processing, Computer-Assisted ,Scanning ,correlative light and electron microscopy ,SBF-SEM ,Enterovirus ,Microscopy ,0303 health sciences ,biology ,Chemistry ,CLEM ,Lipid Droplets/ultrastructure ,QR1-502 ,3. Good health ,Cell biology ,Research Article ,Context (language use) ,replication organelle biogenesis ,Electron ,Microbiology ,Host-Microbe Biology ,03 medical and health sciences ,Virology ,membrane structure ,Organelle ,Enterovirus Infections ,Animals ,Vero Cells ,030304 developmental biology ,coxsackievirus ,030306 microbiology ,Endoplasmic reticulum ,Lipid Droplets ,biology.organism_classification ,picornavirus ,Viral replication ,Ultrastructure ,Microscopy, Electron, Scanning ,serial block-face scanning electron microscopy ,Golgi Apparatus/ultrastructure ,Biogenesis - Abstract
Enteroviruses are causative agents of a range of human diseases. The replication of these viruses within cells relies on specialized membranous structures termed replication organelles (ROs) that form during infection but whose origin remains elusive. To capture the emergence of enterovirus ROs, we use correlative light and serial block-face scanning electron microscopy, a powerful method to pinpoint rare events in their whole-cell ultrastructural context. RO biogenesis was found to occur first at ER and then at Golgi membranes. Extensive contacts were found between early ROs and lipid droplets (LDs), which likely serve to provide LD-derived lipids required for replication. Together, these data establish the dual origin of enterovirus ROs and the chronology of their biogenesis at different supporting cellular membranes., Enterovirus genome replication occurs at virus-induced structures derived from cellular membranes and lipids. However, the origin of these replication organelles (ROs) remains uncertain. Ultrastructural evidence of the membrane donor is lacking, suggesting that the sites of its transition into ROs are rare or fleeting. To overcome this challenge, we combined live-cell imaging and serial block-face scanning electron microscopy of whole cells to capture emerging enterovirus ROs. The first foci of fluorescently labeled viral protein correlated with ROs connected to the endoplasmic reticulum (ER) and preceded the appearance of ROs stemming from the trans-Golgi network. Whole-cell data sets further revealed striking contact regions between ROs and lipid droplets that may represent a route for lipid shuttling to facilitate RO proliferation and genome replication. Our data provide direct evidence that enteroviruses use ER and then Golgi membranes to initiate RO formation, demonstrating the remarkable flexibility with which enteroviruses usurp cellular organelles.
- Published
- 2019
234. The 2nd sialic acid-binding site of influenza A virus neuraminidase is an important determinant of the hemagglutinin-neuraminidase-receptor balance
- Author
-
Du, Wenjuan, Guo, Hongbo, Nijman, Vera S, Doedt, Jennifer, van der Vries, Erhard, van der Lee, Joline, Li, Zeshi, Boons, Geert-Jan, van Kuppeveld, Frank J M, de Vries, Erik, Matrosovich, Mikhail, de Haan, Cornelis A M, LS Virologie, dIRAS RA-1, dI&I I&I-1, Sub Chemical Biology and Drug Discovery, Afd Chemical Biology and Drug Discovery, Chemical Biology and Drug Discovery, LS Virologie, dIRAS RA-1, dI&I I&I-1, Sub Chemical Biology and Drug Discovery, Afd Chemical Biology and Drug Discovery, and Chemical Biology and Drug Discovery
- Subjects
RNA viruses ,medicine.disease_cause ,Biochemistry ,Influenza A Virus, H2N2 Subtype ,Madin Darby Canine Kidney Cells ,Virions ,chemistry.chemical_compound ,Binding Analysis ,Lectins ,Chlorocebus aethiops ,Influenza A virus ,Biology (General) ,Pathology and laboratory medicine ,0303 health sciences ,biology ,Chemistry ,030302 biochemistry & molecular biology ,Eukaryota ,Medical microbiology ,Recombinant Proteins ,Vertebrates ,Viruses ,Receptors, Virus ,Pathogens ,Hemagglutinin-neuraminidase ,Research Article ,Virus genetics ,QH301-705.5 ,Immunology ,Neuraminidase ,Sialic acid binding ,Viral Structure ,Research and Analysis Methods ,Microbiology ,Virus ,Birds ,03 medical and health sciences ,Viral Proteins ,Dogs ,Virology ,Genetics ,medicine ,Animals ,Humans ,Influenza viruses ,Molecular Biology ,Vero Cells ,Receptor Binding Assays ,Chemical Characterization ,030304 developmental biology ,Medicine and health sciences ,Binding Sites ,Influenza A Virus, H3N2 Subtype ,Virion ,Organisms ,Viral pathogens ,Biology and Life Sciences ,Proteins ,RC581-607 ,N-Acetylneuraminic Acid ,Viral Replication ,Sialic acid ,Microbial pathogens ,Viral replication ,Amniotes ,biology.protein ,Parasitology ,Immunologic diseases. Allergy ,Orthomyxoviruses - Abstract
Influenza A virus (IAV) neuraminidase (NA) receptor-destroying activity and hemagglutinin (HA) receptor-binding affinity need to be balanced with the host receptor repertoire for optimal viral fitness. NAs of avian, but not human viruses, contain a functional 2nd sialic acid (SIA)-binding site (2SBS) adjacent to the catalytic site, which contributes to sialidase activity against multivalent substrates. The receptor-binding specificity and potentially crucial contribution of the 2SBS to the HA-NA balance of virus particles is, however, poorly characterized. Here, we elucidated the receptor-binding specificity of the 2SBS of N2 NA and established an important role for this site in the virion HA-NA-receptor balance. NAs of H2N2/1957 pandemic virus with or without a functional 2SBS and viruses containing this NA were analysed. Avian-like N2, with a restored 2SBS due to an amino acid substitution at position 367, was more active than human N2 on multivalent substrates containing α2,3-linked SIAs, corresponding with the pronounced binding-specificity of avian-like N2 for these receptors. When introduced into human viruses, avian-like N2 gave rise to altered plaque morphology and decreased replication compared to human N2. An opposite replication phenotype was observed when N2 was combined with avian-like HA. Specific bio-layer interferometry assays revealed a clear effect of the 2SBS on the dynamic interaction of virus particles with receptors. The absence or presence of a functional 2SBS affected virion-receptor binding and receptor cleavage required for particle movement on a receptor-coated surface and subsequent NA-dependent self-elution. The contribution of the 2SBS to virus-receptor interactions depended on the receptor-binding properties of HA and the identity of the receptors used. We conclude that the 2SBS is an important and underappreciated determinant of the HA-NA-receptor balance. The rapid loss of a functional 2SBS in pandemic viruses may have served to balance the novel host receptor-repertoire and altered receptor-binding properties of the corresponding HA protein., Author summary Influenza A viruses infect birds and mammals. They contain receptor-binding (HA) and receptor-destroying (NA) proteins, which are crucial determinants of host tropism and pathogenesis. It is generally accepted that the functional properties of HA and NA need to be well balanced to enable virion penetration of the receptor-rich mucus layer, binding to host cells, and release of newly assembled particles. This HA-NA-receptor balance is, however, poorly characterized resulting in part from a lack of suitable assays to measure this balance. In addition, NA is much less studied than HA. NA contains, besides its receptor-cleavage site, a 2nd receptor-binding site, which is functional in avian, but not in human viruses. We now show that this 2nd receptor-binding site prefers binding to avian-type receptors and promotes cleavage of substrates carrying this receptor. Furthermore, by using novel assays, we established an important role for this site in the HA-NA-receptor balance of virus particles as it contributes to receptor binding and cleavage by virions, the latter of which is required for virion movement and self-elution from receptors. The results may provide an explanation for the rapid loss of a functional 2nd receptor-binding site in human pandemic viruses.
- Published
- 2019
235. Blocking transmission of Middle East respiratory syndrome coronavirus (MERS-CoV) in llamas by vaccination with a recombinant spike protein
- Author
-
Rodon, Jordi, Okba, Nisreen M.A., Te, Nigeer, van Dieren, Brenda, Bosch, Berend Jan, Bensaid, Albert, Segalés, Joaquim, Haagmans, Bart L., Vergara-Alert, Júlia, LS Virologie, dI&I I&I-1, Virology, LS Virologie, dI&I I&I-1, Producció Animal, and Sanitat Animal
- Subjects
0301 basic medicine ,Camelus ,Virus transmission ,Middle East respiratory syndrome coronavirus ,Epidemiology ,viruses ,030106 microbiology ,Immunology ,S1-protein-based vaccine ,Biology ,medicine.disease_cause ,Antibodies, Viral ,Microbiology ,Virus ,Article ,law.invention ,03 medical and health sciences ,MERS-CoV ,SDG 3 - Good Health and Well-being ,law ,Virology ,Zoonoses ,Drug Discovery ,medicine ,Virus-neutralizing Antibody ,Animals ,Humans ,Animal model ,llama ,Transmission (medicine) ,Vaccination ,Outbreak ,Viral Vaccines ,General Medicine ,virus transmission ,Antibodies, Neutralizing ,3. Good health ,030104 developmental biology ,Infectious Diseases ,Spike Glycoprotein, Coronavirus ,Recombinant DNA ,Parasitology ,Coronavirus Infections ,Camelids, New World - Abstract
The ongoing Middle East respiratory syndrome coronavirus (MERS-CoV) outbreaks pose a worldwide public health threat. Blocking MERS-CoV zoonotic transmission from dromedary camels, the animal reservoir, could potentially reduce the number of primary human cases. Here we report MERS-CoV transmission from experimentally infected llamas to naïve animals. Directly inoculated llamas shed virus for at least 6 days and could infect all in-contact naïve animals 4-5 days after exposure. With the aim to block virus transmission, we examined the efficacy of a recombinant spike S1-protein vaccine. In contrast to naïve animals, in-contact vaccinated llamas did not shed infectious virus upon exposure to directly inoculated llamas, consistent with the induction of strong virus neutralizing antibody responses. Our data provide further evidence that vaccination of the reservoir host may impede MERS-CoV zoonotic transmission to humans. info:eu-repo/semantics/publishedVersion
- Published
- 2019
236. No evidence for viral small RNA production and antiviral function of Argonaute 2 in human cells
- Author
-
Schuster, Susan, Overheul, Gijs J, Bauer, Lisa, van Kuppeveld, Frank J M, van Rij, Ronald P, LS Virologie, dI&I I&I-1, LS Virologie, and dI&I I&I-1
- Subjects
0301 basic medicine ,Small interfering RNA ,Sindbis virus ,Small RNA ,Picornavirus ,viruses ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,lcsh:Medicine ,Biology ,Virus-host interactions ,Virus Replication ,Article ,Virus ,Cell Line ,Viral Proteins ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,Viral Small RNA ,RNA interference ,Cell Line, Tumor ,Humans ,RNA Viruses ,Encephalomyocarditis virus ,RNA, Small Interfering ,lcsh:Science ,Innate immunity ,Multidisciplinary ,030102 biochemistry & molecular biology ,Virus–host interactions ,lcsh:R ,DNA Viruses ,RNA ,biology.organism_classification ,Virology ,Enterovirus A, Human ,Enterovirus B, Human ,3. Good health ,HEK293 Cells ,030104 developmental biology ,RNAi ,Argonaute Proteins ,RNA, Viral ,lcsh:Q ,RNA Interference ,Sindbis Virus ,HeLa Cells - Abstract
RNA interference (RNAi) has strong antiviral activity in a range of animal phyla, but the extent to which RNAi controls virus infection in chordates, and specifically mammals remains incompletely understood. Here we analyze the antiviral activity of RNAi against a number of positive-sense RNA viruses using Argonaute-2 deficient human cells. In line with absence of virus-derived siRNAs, Sindbis virus, yellow fever virus, and encephalomyocarditis virus replicated with similar kinetics in wildtype cells and Argonaute-2 deficient cells. Coxsackievirus B3 (CVB3) carrying mutations in the viral 3A protein, previously proposed to be a virus-encoded suppressor of RNAi in another picornavirus, human enterovirus 71, had a strong replication defect in wildtype cells. However, this defect was not rescued in Argonaute-2 deficient cells, arguing against a role of CVB3 3A as an RNAi suppressor. In agreement, neither infection with wildtype nor 3A mutant CVB3 resulted in small RNA production with the hallmarks of canonical vsiRNAs. Together, our results argue against strong antiviral activity of RNAi under these experimental conditions, but do not exclude that antiviral RNAi may be functional under other cellular, experimental, or physiological conditions in mammals.
- Published
- 2019
237. Pan-European study on the prevalence of the feline leukaemia virus infection - Reported by the european advisory board on cat diseases (ABCD Europe)
- Author
-
Studer, Nadine, Lutz, Hans, Saegerman, Claude, Gönczi, Enikö, Meli, Marina L, Boo, Gianluca, Hartmann, Katrin, Hosie, Margaret J, Moestl, Karin, Tasker, Séverine, Belák, Sándor, Lloret, Albert, Boucraut-Baralon, Corine, Egberink, Herman F, Pennisi, Maria-Grazia, Truyen, Uwe, Frymus, Tadeusz, Thiry, Etienne, Marsilio, Fulvio, Addie, Diane, Hochleithner, Manfred, Tkalec, Filip, Vizi, Zsuzsanna, Brunetti, Anna, Georgiev, Boyko, Ludwig-Begall, Louisa F, Tschuor, Flurin, Mooney, Carmel T, Eliasson, Catarina, Orro, Janne, Johansen, Helle, Juuti, Kirsi, Krampl, Igor, Kovalenko, Kaspars, Šengaut, Jakov, Sobral, Cristina, Borska, Petra, Kovaříková, Simona, Hofmann-Lehmann, Regina, LS Klinisch Onderzoek Wagenaar, LS Virologie, dI&I I&I-1, dI&I I&I-4, LS Klinisch Onderzoek Wagenaar, LS Virologie, dI&I I&I-1, dI&I I&I-4, University of Zurich, and Hofmann-Lehmann, Regina
- Subjects
Male ,0301 basic medicine ,FeLV ,animal diseases ,viruses ,lcsh:QR1-502 ,Prevalence ,Leucèmia felina ,Cat Diseases ,lcsh:Microbiology ,0403 veterinary science ,hemic and lymphatic diseases ,Epidemiology ,Medicine ,risk factors ,Prospective Studies ,11434 Center for Clinical Studies ,FeLV, retrovirus, prevalence, risk factors, protective factors, RT-qPCR, virus shedding, vaccination, gross domestic product at purchasing power parity per capita, veterinary sciences ,Stomatitis ,CATS ,Retrovirus ,Leukemia Virus, Feline ,Incidence (epidemiology) ,Vaccination ,virus diseases ,Virus shedding ,04 agricultural and veterinary sciences ,Europe ,retrovirus ,qPCR ,Infectious Diseases ,11404 Department of Clinical Diagnostics and Services ,Patogènesi ,Female ,gross domestic product at purchasing power parity per capita ,prevalence ,protective factors ,RT-qPCR ,vaccination ,veterinary sciences ,virus shedding ,medicine.symptom ,medicine.medical_specialty ,040301 veterinary sciences ,610 Medicine & health ,Anorexia ,Article ,RT ,03 medical and health sciences ,Virology ,Internal medicine ,Gross domestic product at purchasing power parity per capita ,Animals ,Viremia ,Viral shedding ,Saliva ,business.industry ,2725 Infectious Diseases ,medicine.disease ,Tumor Virus Infections ,Gats ,030104 developmental biology ,Protective factors ,Risk factors ,Cats ,2406 Virology ,business ,Retroviridae Infections - Abstract
Feline leukaemia virus (FeLV) is a retrovirus associated with fatal disease in progressively infected cats. While testing/removal and vaccination led to a decreased prevalence of FeLV, recently, this decrease has reportedly stagnated in some countries. This study aimed to prospectively determine the prevalence of FeLV viraemia in cats taken to veterinary facilities in 32 European countries. FeLV viral RNA was semiquantitatively detected in saliva, using RT-qPCR as a measure of viraemia. Risk and protective factors were assessed using an online questionnaire to report geographic, demographic, husbandry, FeLV vaccination, and clinical data. The overall prevalence of FeLV viraemia in cats visiting a veterinary facility, of which 10.4% were shelter and rescue cats, was 2.3% (141/6005, 95% CI: 2.0%&ndash, 2.8%) with the highest prevalences in Portugal, Hungary, and Italy/Malta (5.7%&ndash, 8.8%). Using multivariate analysis, seven risk factors (Southern Europe, male intact, 1&ndash, 6 years of age, indoor and outdoor or outdoor-only living, living in a group of &ge, 5 cats, illness), and three protective factors (Northern Europe, Western Europe, pedigree cats) were identified. Using classification and regression tree (CART) analysis, the origin of cats in Europe, pedigree, and access to outdoors were important predictors of FeLV status. FeLV-infected sick cats shed more viral RNA than FeLV-infected healthy cats, and they suffered more frequently from anaemia, anorexia, and gingivitis/stomatitis than uninfected sick cats. Most cats had never been FeLV-vaccinated, vaccination rates were indirectly associated with the gross domestic product (GDP) per capita. In conclusion, we identified countries where FeLV was undetectable, demonstrating that the infection can be eradicated and highlighting those regions where awareness and prevention should be increased.
- Published
- 2019
238. Role of enhanced receptor engagement in the evolution of a pandemic acute hemorrhagic conjunctivitis virus
- Author
-
LS Virologie, Virologie, dI&I I&I-1, Baggen, Jim, Hurdiss, Daniel L, Zocher, Georg, Mistry, Nitesh, Roberts, Richard W, Slager, Jasper J, Guo, Hongbo, van Vliet, Arno L W, Wahedi, Maryam, Benschop, Kimberley, Duizer, Erwin, de Haan, Cornelis A M, de Vries, Erik, Casasnovas, José M, de Groot, Raoul J, Arnberg, Niklas, Stehle, Thilo, Ranson, Neil A, Thibaut, Hendrik Jan, van Kuppeveld, Frank J M, LS Virologie, Virologie, dI&I I&I-1, Baggen, Jim, Hurdiss, Daniel L, Zocher, Georg, Mistry, Nitesh, Roberts, Richard W, Slager, Jasper J, Guo, Hongbo, van Vliet, Arno L W, Wahedi, Maryam, Benschop, Kimberley, Duizer, Erwin, de Haan, Cornelis A M, de Vries, Erik, Casasnovas, José M, de Groot, Raoul J, Arnberg, Niklas, Stehle, Thilo, Ranson, Neil A, Thibaut, Hendrik Jan, and van Kuppeveld, Frank J M
- Published
- 2018
239. Virus detection in high-throughput sequencing data without a reference genome of the host
- Author
-
Kruppa, Jochen, Jo, Wendy K, van der Vries, Erhard, Ludlow, Martin, Osterhaus, Albert, Baumgaertner, Wolfgang, Jung, Klaus, LS Virologie, dIRAS RA-1, dI&I I&I-1, LS Virologie, dIRAS RA-1, and dI&I I&I-1
- Subjects
0301 basic medicine ,Microbiology (medical) ,Databases, Pharmaceutical ,Sequence assembly ,Genome, Viral ,Computational biology ,Biology ,Microbiology ,Genome ,Business process discovery ,03 medical and health sciences ,Genetics ,Animals ,Humans ,Amino Acid Sequence ,Virus discovery ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Base Sequence ,030102 biochemistry & molecular biology ,Computational Biology ,High-Throughput Nucleotide Sequencing ,Genomics ,Pipeline (software) ,Decoy database ,Pipeline transport ,030104 developmental biology ,Infectious Diseases ,Virus Diseases ,Metagenomics ,Viruses ,Read mapping ,Reference genome ,Host (network) ,Algorithms - Abstract
Discovery of novel viruses in host samples is a multidisciplinary process which relies increasingly on next-generation sequencing (NGS) followed by computational analysis. A crucial step in this analysis is to separate host sequence reads from the sequence reads of the virus to be discovered. This becomes especially difficult if no reference genome of the host is available. Furthermore, if the total number of viral reads in a sample is low, de novo assembly of a virus which is a requirement for most existing pipelines is hard to realize. We present a new modular, computational pipeline for discovery of novel viruses in host samples. While existing pipelines rely on the availability of the hosts reference genome for filtering sequence reads, our new pipeline can also cope with cases for which no reference genome is available. As a further novelty of our method a decoy module is used to assess false classification rates in the discovery process. Additionally, viruses with a low read coverage can be identified and visually reviewed. We validate our pipeline on simulated data as well as two experimental samples with known virus content. For the experimental samples, we were able to reproduce the laboratory findings. Our newly developed pipeline is applicable for virus detection in a wide range of host species. The three modules we present can either be incorporated individually in other pipelines or be used as a stand-alone pipeline. We are the first to present a decoy approach within a virus detection pipeline that can be used to assess error rates so that the quality of the final result can be judged. We provide an implementation of our modules via Github. However, the principle of the modules can easily be re-implemented by other researchers.
- Published
- 2018
240. Les Journées Francophones de Virologie, une dix-septième édition marquée du sceau de la riche diversité de notre discipline
- Author
-
Francis, Barin, Astrid, Vabret, Etienne, Thiry, Noël, Tordo, Yves, Gaudin, Denis, Gerlier, and Rédacteurs de Virologie
- Published
- 2020
241. [Hurry up, a vaccine!]
- Author
-
Le Comité de Crise Covid-de la Société Française de Virologie
- Subjects
Pediatrics ,medicine.medical_specialty ,Vaccines ,Infectious Diseases ,business.industry ,Virology ,medicine ,MEDLINE ,business - Published
- 2020
242. [Chloroquine antiviral potential: 130 years old and still active?]
- Author
-
Le Comité de Crise Covid-de la Société Française de Virologie
- Published
- 2020
243. Dynamics in the murine norovirus capsid revealed by high-resolution cryo-EM
- Author
-
Snowden, Joseph S., Chiu, Wah, Hurdiss, Daniel L., Adeyemi, Oluwapelumi O., Ranson, Neil A., Herod, Morgan R., Stonehouse, Nicola J., dI&I I&I-1, and Virologie
- Subjects
0301 basic medicine ,RNA viruses ,Models, Molecular ,Viral Diseases ,Hot Temperature ,Cryo-electron microscopy ,viruses ,ved/biology.organism_classification_rank.species ,High resolution ,medicine.disease_cause ,Pathology and Laboratory Medicine ,Viral Packaging ,Virions ,Mice ,0302 clinical medicine ,Medicine and Health Sciences ,Electron Microscopy ,Biology (General) ,Materials ,Infectivity ,Microscopy ,General Neuroscience ,3. Good health ,Chemistry ,Infectious Diseases ,Capsid ,Medical Microbiology ,Viral Pathogens ,Viruses ,Physical Sciences ,Pathogens ,General Agricultural and Biological Sciences ,Dimerization ,Research Article ,QH301-705.5 ,Icosahedral symmetry ,Materials Science ,Computational biology ,Biology ,Viral Structure ,Research and Analysis Methods ,Microbiology ,General Biochemistry, Genetics and Molecular Biology ,Caliciviruses ,03 medical and health sciences ,Structure-Activity Relationship ,Extraction techniques ,Viral life cycle ,Protein Domains ,Virology ,medicine ,Animals ,Dimers ,Microbial Pathogens ,General Immunology and Microbiology ,Biology and life sciences ,ved/biology ,Norovirus ,Cryoelectron Microscopy ,Organisms ,Calicivirus Infection ,Electron Cryo-Microscopy ,Polymer Chemistry ,Viral Replication ,RNA extraction ,030104 developmental biology ,RAW 264.7 Cells ,Oligomers ,Mutation ,Capsid Proteins ,030217 neurology & neurosurgery ,Murine norovirus - Abstract
Icosahedral viral capsids must undergo conformational rearrangements to coordinate essential processes during the viral life cycle. Capturing such conformational flexibility has been technically challenging yet could be key for developing rational therapeutic agents to combat infections. Noroviruses are nonenveloped, icosahedral viruses of global importance to human health. They are a common cause of acute gastroenteritis, yet no vaccines or specific antiviral agents are available. Here, we use genetics and cryo-electron microscopy (cryo-EM) to study the high-resolution solution structures of murine norovirus as a model for human viruses. By comparing our 3 structures (at 2.9- to 3.1-Å resolution), we show that whilst there is little change to the shell domain of the capsid, the radiating protruding domains are flexible, adopting distinct states both independently and synchronously. In doing so, the capsids sample a range of conformational space, with implications for maintaining virion stability and infectivity., High-resolution cryo-EM reveals that the norovirus capsid continuously samples conformational space, challenging the model of conformational changes in virus structure being orchestrated along linear, irreversible pathways of assembly and disassembly, with implications for infectivity and immune evasion.
- Published
- 2020
244. Viral evasion of cellular stress responses
- Author
-
Rabouw, Huibert Hendrik, LS Virologie, van Kuppeveld, Frank, de Groot, Raoul, Langereis, Martijn, and University Utrecht
- Subjects
Coronavirus ,Translation ,ISRIB ,eIF2B ,Picornavirus ,Antagonist ,eIF2 ,PKR ,ISR ,Integrated Stress Response ,Virus - Abstract
For many viruses, efficient replication is contingent on the ability to evade antiviral responses such as the ISR. Yet, for most viruses it remains poorly understood by what mechanisms the ISR is suppressed. In this thesis we set out to identify new ISR antagonists encoded by picorna- and coronaviruses, and on the elucidation of their molecular mode of action. This will increase our knowledge about evasion mechanisms employed by viruses to protect themselves against antiviral responses. From a cell biological perspective, the study of viral ISR antagonists may also teach us more about how the ISR regulates translation, one of the core processes in life. In chapter 2, we set out to investigate the function of an accessory protein (p4a) encoded by Middle-East Respiratory Syndrome coronavirus (MERS-CoV) as inhibitor of PKR-mediated ISR activation(91). We identify MERS-CoV p4a as a PKR-specific ISR antagonist that functions via dsRNA binding. Consistent with its mode of action of dsRNA sequestration, p4a also prevents the activation of another dsRNA-activated antiviral pathway, the type I IFN induction pathway. While the mode of action of p4a was described before for several viral dsRNA binding proteins, p4a is the first coronavirus protein identified with this function. In chapter 3, we assess the function of a small-molecule inhibitor of the ISR (ISRIB). We observed that ISRIB prevents ISR activation only early in infection, but failed to do so at later time points. Further investigation taught us that ISRIB prevents ISR activity only under conditions of mild stress, as defined by a limited level of eIF2 phosphorylation. High levels of p-eIF2 however may inhibit translation and enhance expression of ISR-induced proteins, even in the presence of ISRIB. Thus, ISRIB makes cells less sensitive rather than insensitive to ISR signaling. These findings are consistent with the previously reported mode of action of ISRIB, and may explain the remarkable absence of side-effects of an ISRIB treatment in vivo. In chapter 4, we investigated the function(s) of an accessory protein, AcP10, encoded by Beluga whale coronavirus. AcP10 prevents p-eIF2 from binding its target eIF2B, and thereby AcP10 ensures that eIF2B remains active even in the presence of p-eIF2. It does so via a direct interaction with eIF2B, likely at an area that overlaps the interaction site for p-eIF2, but not for native eIF2. This mode-of-action was not previously described for any viral or host cell protein, and thus AcP10 is a prototype of an entirely novel class of ISR antagonists. We classified AcP10 as a Class 4 antagonist. In chapter 5, we describe another Class 4 antagonist, the Leader protein encoded by the picornavirus Aichivirus (AiVL). AiVL, like AcP10, binds eIF2B to prevent subsequent association of p-eIF2, thereby rescuing translation in the presence of (high levels) of p-eIF2 irrespective of the eIF2 kinase involved. Yet, AiVL has no homology to AcP10. Thus, Class 4 ISR antagonists arose at least twice, in phylogenetically distinct virus families, by convergent evolution.
- Published
- 2020
245. Dynamics in the murine norovirus capsid revealed by high-resolution cryo-EM
- Author
-
dI&I I&I-1, Virologie, Snowden, Joseph S., Chiu, Wah, Hurdiss, Daniel L., Adeyemi, Oluwapelumi O., Ranson, Neil A., Herod, Morgan R., Stonehouse, Nicola J., dI&I I&I-1, Virologie, Snowden, Joseph S., Chiu, Wah, Hurdiss, Daniel L., Adeyemi, Oluwapelumi O., Ranson, Neil A., Herod, Morgan R., and Stonehouse, Nicola J.
- Published
- 2020
246. Possible absence of trimeron correlations above the Verwey temperature in Fe3 O4
- Author
-
Sub Inorganic Chemistry and Catalysis, LS Virologie, Inorganic Chemistry and Catalysis, Elnaggar, H., Wang, R., Lafuerza, S., Paris, E., Komarek, A. C., Guo, H., Tseng, Y., McNally, D., Frati, F., Haverkort, M. W., Sikora, M., Schmitt, T., De Groot, F. M.F., Sub Inorganic Chemistry and Catalysis, LS Virologie, Inorganic Chemistry and Catalysis, Elnaggar, H., Wang, R., Lafuerza, S., Paris, E., Komarek, A. C., Guo, H., Tseng, Y., McNally, D., Frati, F., Haverkort, M. W., Sikora, M., Schmitt, T., and De Groot, F. M.F.
- Published
- 2020
247. Encephalitozoon cuniculi infection in cats: European guidelines from the ABCD on prevention and management
- Author
-
Klinische infectiologie en microb. lab., Virologie, dI&I I&I-1, dI&I I&I-4, Addie, Diane D., Tasker, Séverine, Boucraut-Baralon, Corine, Belák, Sandor, Egberink, Herman, Frymus, Tadeusz, Hartmann, Katrin, Hofmann-Lehmann, Regina, Marsilio, Fulvio, Lloret, Albert, Pennisi, Maria Grazia, Thiry, Etienne, Truyen, Uwe, Hosie, Margaret J., Möstl, Karin, Klinische infectiologie en microb. lab., Virologie, dI&I I&I-1, dI&I I&I-4, Addie, Diane D., Tasker, Séverine, Boucraut-Baralon, Corine, Belák, Sandor, Egberink, Herman, Frymus, Tadeusz, Hartmann, Katrin, Hofmann-Lehmann, Regina, Marsilio, Fulvio, Lloret, Albert, Pennisi, Maria Grazia, Thiry, Etienne, Truyen, Uwe, Hosie, Margaret J., and Möstl, Karin
- Published
- 2020
248. Accurate serology for SARS-CoV-2 and common human coronaviruses using a multiplex approach
- Author
-
Virologie, dI&I I&I-1, van Tol, Sophie, Mögling, Ramona, Li, Wentao, Godeke, Gert-Jan, Swart, Arno, Bergmans, Barbara, Brandenburg, Afke, Kremer, Kristin, Murk, Jean-Luc, van Beek, Josine, Wintermans, Bas, Reimerink, Johan, Bosch, Berend-Jan, Reusken, Chantal, Virologie, dI&I I&I-1, van Tol, Sophie, Mögling, Ramona, Li, Wentao, Godeke, Gert-Jan, Swart, Arno, Bergmans, Barbara, Brandenburg, Afke, Kremer, Kristin, Murk, Jean-Luc, van Beek, Josine, Wintermans, Bas, Reimerink, Johan, Bosch, Berend-Jan, and Reusken, Chantal
- Published
- 2020
249. Development of a SARS-CoV-2 Total Antibody Assay and the Dynamics of Antibody Response over Time in Hospitalized and Nonhospitalized Patients with COVID-19
- Author
-
Virologie, dI&I I&I-1, Amsterdam University Medical Center COVID-19 Biobank Study Group, Virologie, dI&I I&I-1, and Amsterdam University Medical Center COVID-19 Biobank Study Group
- Published
- 2020
250. Older adults lack SARS CoV-2 cross-reactive T lymphocytes directed to human coronaviruses OC43 and NL63
- Author
-
Virologie, dI&I I&I-1, Bedrijfsvoering, Saletti, Giulietta, Gerlach, Thomas, Jansen, Janina M, Molle, Antonia, Elbahesh, Husni, Ludlow, Martin, Li, Wentao, Bosch, Berend-Jan, Osterhaus, Albert D M E, Rimmelzwaan, Guus F, Virologie, dI&I I&I-1, Bedrijfsvoering, Saletti, Giulietta, Gerlach, Thomas, Jansen, Janina M, Molle, Antonia, Elbahesh, Husni, Ludlow, Martin, Li, Wentao, Bosch, Berend-Jan, Osterhaus, Albert D M E, and Rimmelzwaan, Guus F
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.