61 results on '"Zahra Fagrouch"'
Search Results
2. A low dose of RBD and TLR7/8 agonist displayed on influenza virosome particles protects rhesus macaque against SARS-CoV-2 challenge
- Author
-
Gerrit Koopman, Mario Amacker, Toon Stegmann, Ernst J. Verschoor, Babs E. Verstrepen, Farien Bhoelan, Denzel Bemelman, Kinga P. Böszörményi, Zahra Fagrouch, Gwendoline Kiemenyi-Kayere, Daniella Mortier, Dagmar E. Verel, Henk Niphuis, Roja Fidel Acar, Ivanela Kondova, Yolanda S. Kap, Willy M. J. M. Bogers, Petra Mooij, and Sylvain Fleury
- Subjects
Medicine ,Science - Abstract
Abstract Influenza virosomes serve as antigen delivery vehicles and pre-existing immunity toward influenza improves the immune responses toward antigens. Here, vaccine efficacy was evaluated in non-human primates with a COVID-19 virosome-based vaccine containing a low dose of RBD protein (15 µg) and the adjuvant 3M-052 (1 µg), displayed together on virosomes. Vaccinated animals (n = 6) received two intramuscular administrations at week 0 and 4 and challenged with SARS-CoV-2 at week 8, together with unvaccinated control animals (n = 4). The vaccine was safe and well tolerated and serum RBD IgG antibodies were induced in all animals and in the nasal washes and bronchoalveolar lavages in the three youngest animals. All control animals became strongly sgRNA positive in BAL, while all vaccinated animals were protected, although the oldest vaccinated animal (V1) was transiently weakly positive. The three youngest animals had also no detectable sgRNA in nasal wash and throat. Cross-strain serum neutralizing antibodies toward Wuhan-like, Alpha, Beta, and Delta viruses were observed in animals with the highest serum titers. Pro-inflammatory cytokines IL-8, CXCL-10 and IL-6 were increased in BALs of infected control animals but not in vaccinated animals. Virosomes-RBD/3M-052 prevented severe SARS-CoV-2, as shown by a lower total lung inflammatory pathology score than control animals.
- Published
- 2023
- Full Text
- View/download PDF
3. Vaccine-induced neutralizing antibody responses to seasonal influenza virus H1N1 strains are not enhanced during subsequent pandemic H1N1 infection
- Author
-
Petra Mooij, Daniella Mortier, Aafke Aartse, Alexandre B. Murad, Ricardo Correia, António Roldão, Paula M. Alves, Zahra Fagrouch, Dirk Eggink, Norbert Stockhofe, Othmar G. Engelhardt, Ernst J. Verschoor, Marit J. van Gils, Willy M. Bogers, Manuel J. T. Carrondo, Edmond J. Remarque, and Gerrit Koopman
- Subjects
influenza ,original antigenic sin ,back-boosting ,non-human primates ,antibody response ,primary response ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The first exposure to influenza is presumed to shape the B-cell antibody repertoire, leading to preferential enhancement of the initially formed responses during subsequent exposure to viral variants. Here, we investigated whether this principle remains applicable when there are large genetic and antigenic differences between primary and secondary influenza virus antigens. Because humans usually have a complex history of influenza virus exposure, we conducted this investigation in influenza-naive cynomolgus macaques. Two groups of six macaques were immunized four times with influenza virus-like particles (VLPs) displaying either one (monovalent) or five (pentavalent) different hemagglutinin (HA) antigens derived from seasonal H1N1 (H1N1) strains. Four weeks after the final immunization, animals were challenged with pandemic H1N1 (H1N1pdm09). Although immunization resulted in robust virus-neutralizing responses to all VLP-based vaccine strains, there were no cross-neutralization responses to H1N1pdm09, and all animals became infected. No reductions in viral load in the nose or throat were detected in either vaccine group. After infection, strong virus-neutralizing responses to H1N1pdm09 were induced. However, there were no increases in virus-neutralizing titers against four of the five H1N1 vaccine strains; and only a mild increase was observed in virus-neutralizing titer against the influenza A/Texas/36/91 vaccine strain. After H1N1pdm09 infection, both vaccine groups showed higher virus-neutralizing titers against two H1N1 strains of intermediate antigenic distance between the H1N1 vaccine strains and H1N1pdm09, compared with the naive control group. Furthermore, both vaccine groups had higher HA-stem antibodies early after infection than the control group. In conclusion, immunization with VLPs displaying HA from antigenically distinct H1N1 variants increased the breadth of the immune response during subsequent H1N1pdm09 challenge, although this phenomenon was limited to intermediate antigenic variants.
- Published
- 2023
- Full Text
- View/download PDF
4. Safety and immunogenicity of four-segmented Rift Valley fever virus in the common marmoset
- Author
-
Paul J. Wichgers Schreur, Petra Mooij, Gerrit Koopman, Babs E. Verstrepen, Zahra Fagrouch, Daniella Mortier, Nikki van Driel, Jet Kant, Sandra van de Water, Willy M. Bogers, Carine Punt, Lucien van Keulen, Ernst J. Verschoor, and Jeroen Kortekaas
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Rift Valley fever virus (RVFV) is an emerging mosquito-borne bunyavirus that is highly pathogenic to wild and domesticated ruminants, camelids, and humans. While animals are exclusively infected via mosquito bites, humans can also be infected via contact with contaminated tissues or blood. No human vaccine is available and commercialized veterinary vaccines do not optimally combine efficacy with safety. We previously reported the development of two novel live-attenuated RVF vaccines, created by splitting the M genome segment and deleting the major virulence determinant NSs. The vaccine candidates, referred to as the veterinary vaccine vRVFV-4s and the human vaccine hRVFV-4s, were shown to induce protective immunity in multiple species after a single vaccination. Anticipating accidental exposure of humans to the veterinary vaccine and the application of hRVFV-4s to humans, the safety of each vaccine was evaluated in the most susceptible nonhuman primate model, the common marmoset (Callithrix jacchus). Marmosets were inoculated with high doses of each vaccine and were monitored for clinical signs as well as for vaccine virus dissemination, shedding, and spreading to the environment. To accurately assess the attenuation of both vaccine viruses, separate groups of marmosets were inoculated with the parent wild-type RVFV strains. Both wild-type strains induced high viremia and disseminated to primary target organs, associated with mild-to-severe morbidity. In contrast, both vaccines were well tolerated with no evidence of dissemination and shedding while inducing potent neutralizing antibody responses. The results of the studies support the unprecedented safety profile of both vaccines for animals and humans.
- Published
- 2022
- Full Text
- View/download PDF
5. Poxvirus MVA Expressing SARS-CoV-2 S Protein Induces Robust Immunity and Protects Rhesus Macaques From SARS-CoV-2
- Author
-
Petra Mooij, Juan García-Arriaza, Patricia Pérez, Adrian Lázaro-Frías, Babs E. Verstrepen, Kinga P. Böszörményi, Daniella Mortier, Zahra Fagrouch, Gwendoline Kiemenyi-Kayere, Henk Niphuis, Roja Fidel Acar, Lisette Meijer, Marieke A. Stammes, Ivanela Kondova, Ernst J. Verschoor, Corine H. GeurtsvanKessel, Erwin de Bruin, Reina S. Sikkema, Joanna Luczkowiak, Rafael Delgado, Dolores Montenegro, Eugenia Puentes, Esteban Rodríguez, Willy M. J. M. Bogers, Gerrit Koopman, and Mariano Esteban
- Subjects
SARS-CoV-2 ,COVID-19 ,spike ,MVA vaccine ,rhesus macaques ,safety ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Novel safe, immunogenic, and effective vaccines are needed to control the COVID-19 pandemic, caused by SARS-CoV-2. Here, we describe the safety, robust immunogenicity, and potent efficacy elicited in rhesus macaques by a modified vaccinia virus Ankara (MVA) vector expressing a full-length SARS-CoV-2 spike (S) protein (MVA-S). MVA-S vaccination was well tolerated and induced S and receptor-binding domain (RBD)-binding IgG antibodies and neutralizing antibodies against SARS-CoV-2 and several variants of concern. S-specific IFNγ, but not IL-4, -producing cells were also elicited. After SARS-CoV-2 challenge, vaccinated animals showed a significant strong reduction of virus loads in bronchoalveolar lavages (BAL) and decreased levels in throat and nasal mucosa. Remarkably, MVA-S also protected macaques from fever and infection-induced cytokine storm. Computed tomography and histological examination of the lungs showed reduced lung pathology in MVA-S-vaccinated animals. These findings favor the use of MVA-S as a potential vaccine for SARS-CoV-2 in clinical trials.
- Published
- 2022
- Full Text
- View/download PDF
6. Pinpointing the PRDM9-PRDM7 Gene Duplication Event During Primate Divergence
- Author
-
Sacha Heerschop, Zahra Fagrouch, Ernst J. Verschoor, and Hans Zischler
- Subjects
PRDM7 ,PRDM9 ,gene duplication ,paralogization ,primate evolution ,Genetics ,QH426-470 - Abstract
Studies on the function of PRDM9 in model systems and its evolution during vertebrate divergence shed light on the basic molecular mechanisms of hybrid sterility and its evolutionary consequences. However, information regarding PRDM9-homolog, PRDM7, whose origin is placed in the primate evolutionary tree, as well as information about the fast-evolving DNA-binding zinc finger array of strepsirrhine PRDM9 are scarce. Thus, we aimed to narrow down the date of the duplication event leading to the emergence of PRDM7 during primate evolution by comparing the phylogenetic tree reconstructions of representative primate samples of PRDM orthologs and paralogs. To confirm our PRDM7 paralogization pattern, database-deposited sequences were used to test the presence/absence patterns expected from the paralogization timing. In addition, we extended the existing phylogenetic tree of haplorrhine PRDM9 zinc fingers with their strepsirrhine counterparts. The inclusion of strepsirrhine zinc fingers completes the PRDM9 primate phylogeny. Moreover, the updated phylogeny of PRDM9 zinc fingers showed distinct clusters of strepsirrhine, tarsier, and anthropoid degenerated zinc fingers. Here, we show that PRDM7 emerged on the branch leading to the most recent common ancestor of catarrhines; therefore, its origin is more recent than previously expected. A more detailed character evolutionary study suggests that PRDM7 may have evolved differently in Cercopithecoidea as compared to Hominoidea: it lacks the first four exons in Old World monkeys orthologs and exon 10 in Papionini orthologs. Dating the origin of PRDM7 is essential for further studies investigating why Hominoidea representatives need another putative histone methyltransferase in the testis.
- Published
- 2021
- Full Text
- View/download PDF
7. Bronchoalveolar lavage affects thorax computed tomography of healthy and SARS-CoV-2 infected rhesus macaques (Macaca mulatta).
- Author
-
Annemiek Maaskant, Lisette Meijer, Zahra Fagrouch, Jaco Bakker, Leo van Geest, Dian G M Zijlmans, Babs E Verstrepen, Jan A M Langermans, Ernst J Verschoor, and Marieke A Stammes
- Subjects
Medicine ,Science - Abstract
Medical imaging as method to assess the longitudinal process of a SARS-CoV-2 infection in non-human primates is commonly used in research settings. Bronchoalveolar lavage (BAL) is regularly used to determine the local virus production and immune effects of SARS-CoV-2 in the lower respiratory tract. However, the potential interference of those two diagnostic modalities is unknown in non-human primates. The current study investigated the effect and duration of BAL on computed tomography (CT) in both healthy and experimentally SARS-CoV-2-infected female rhesus macaques (Macaca mulatta). In addition, the effect of subsequent BALs was reviewed. Thorax CTs and BALs were obtained from four healthy animals and 11 experimentally SARS-CoV-2-infected animals. From all animals, CTs were obtained just before BAL, and 24 hours post-BAL. Additionally, from the healthy animals, CTs immediately after, and four hours post-BAL were obtained. Thorax CTs were evaluated for alterations in lung density, measured in Hounsfield units, and a visual semi-quantitative scoring system. An increase in the lung density was observed on the immediately post-BAL CT but resolved within 24 hours in the healthy animals. In the infected animals, a significant difference in both the lung density and CT score was still found 24 hours after BAL. Furthermore, the differences between time points in CT score were increased for the second BAL. These results indicate that the effect of BAL on infected lungs is not resolved within the first 24 hours. Therefore, it is important to acknowledge the interference between BAL and CT in rhesus macaques.
- Published
- 2021
- Full Text
- View/download PDF
8. TB and SIV Coinfection; a Model for Evaluating Vaccine Strategies against TB Reactivation in Asian Origin Cynomolgus Macaques: A Pilot Study Using BCG Vaccination
- Author
-
Andrew D. White, Laura Sibley, Jennie Gullick, Charlotte Sarfas, Simon Clark, Zahra Fagrouch, Ernst Verschoor, Francisco J. Salguero, Mike Dennis, and Sally Sharpe
- Subjects
tuberculosis ,SIV ,macaques ,coinfection ,reactivation ,Medicine - Abstract
This pilot study aimed to determine the utility of a cynomolgus macaque model of coinfection with simian immunodeficiency virus (SIV) for the assessment of vaccines designed to prevent reactivation of TB. Following infection caused by aerosol exposure to an ultralow dose of Mycobacterium tuberculosis (M. tb), data trends indicated that subsequent coinfection with SIVmac32H perturbed control of M. tb infection as evidenced by the increased occurrence of progressive disease in this group, higher levels of pathology and increased frequency of progressive tuberculous granulomas in the lung. BCG vaccination led to improved control of TB-induced disease and lower viral load in comparison to unvaccinated coinfected animals. The M. tb-specific IFNγ response after exposure to M. tb, previously shown to be associated with bacterial burden, was lower in the BCG-vaccinated group than in the unvaccinated groups. Levels of CD4+ and CD8+ T cells decreased in coinfected animals, with counts recovering more quickly in the BCG-vaccinated group. This pilot study provides proof of concept to support the use of the model for evaluation of interventions against reactivated/exacerbated TB caused by human immunodeficiency virus (HIV) infection.
- Published
- 2021
- Full Text
- View/download PDF
9. Absence of accessory genes in a divergent simian T-lymphotropic virus type 1 isolated from a bonnet macaque (Macaca radiata).
- Author
-
Philippe V Afonso, Zahra Fagrouch, Martin Deijs, Henk Niphuis, Willy Bogers, Antoine Gessain, Lia van der Hoek, and Ernst J Verschoor
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
BackgroundPrimate T-lymphotropic viruses type 1 (PTLV-1) are complex retroviruses infecting both human (HTLV-1) and simian (STLV-1) hosts. They share common epidemiological, clinical and molecular features. In addition to the canonical gag, pol, env retroviral genes, PTLV-1 purportedly encodes regulatory (i.e. Tax, Rex, and HBZ) and accessory proteins (i.e. P12/8, P13, P30). The latter have been found essential for viral persistence in vivo.Methodology/principal findingsWe have isolated a STLV-1 virus from a bonnet macaque (Macaca radiata-Mra18C9), a monkey from India. The complete sequence was obtained and phylogenetic analyses were performed. The Mra18C9 strain is highly divergent from the known PTLV-1 strains. Intriguingly, the Mra18C9 lacks the 3 accessory open reading frames. In order to determine if the absence of accessory proteins is specific to this particular strain, a comprehensive analysis of the complete PTLV-1 genomes available in Genbank was performed and found that the lack of one or many accessory ORF is common among PTLV-1.ConclusionThis study raises many questions regarding the actual nature, role and importance of accessory proteins in the PTLV-1 biology.
- Published
- 2019
- Full Text
- View/download PDF
10. A Bacterially-Expressed Recombinant Envelope Protein from Usutu Virus Induces Neutralizing Antibodies in Rabbits
- Author
-
Kinga Böszörményi, Janet Hirsch, Gwendoline Kiemenyi Kayere, Zahra Fagrouch, Nicole Heijmans, Roberto Rodriguez Garcia, Soesjiel Dwarka, Amy van Dijke, Boyd Aaldijk, Ronald Limpens, Montserrat Barcena, Bram Koster, Babs Verstrepen, Willy Bogers, Clemens Kocken, Gesine Cornellissen, Ernst Verschoor, and Bart Faber
- Subjects
Usutu virus ,USUV ,vaccine development ,flavivirus ,emerging disease ,recombinant E protein ,Medicine - Abstract
Background: Recently, an emerging flavivirus, Usutu virus (USUV), has caused an epidemic among birds in Europe, resulting in a massive die-off in Eurasian blackbirds. Currently found only in Europe and Africa, it can be envisioned that Usutu virus will follow the path of other flaviviruses, like West Nile virus and Zika virus, and will spread via its mosquito vectors and bird hosts to other parts of the world. Several cases of human infections by Usutu virus have already been published. Anticipating this spread, development of an efficacious vaccine would be highly desirable. Method: This study describes the production in E. coli, purification, and refolding of a partial USUV envelope protein. Prior to immunization, the protein was characterized using size exclusion chromatography, transmission electron microscopy and dynamic light scattering, showing the limited presence of virus-like structures, indicating that the protein solution is probably a mixture of mono and multimeric envelope proteins. Results: Immunizations of two rabbits with the refolded E-protein fraction, mixed with a strong adjuvant, resulted in the generation of neutralizing antibodies, as evidenced in an in vitro assay. Discussion: The way forward towards a subunit vaccine against Usutu virus infection is discussed.
- Published
- 2021
- Full Text
- View/download PDF
11. Aerosolized Exposure to H5N1 Influenza Virus Causes Less Severe Disease Than Infection via Combined Intrabronchial, Oral, and Nasal Inoculation in Cynomolgus Macaques
- Author
-
Petra Mooij, Marieke A. Stammes, Daniella Mortier, Zahra Fagrouch, Nikki van Driel, Ernst J. Verschoor, Ivanela Kondova, Willy M. J. M. Bogers, and Gerrit Koopman
- Subjects
influenza ,H5N1 ,infection ,aerosol ,macaques ,Microbiology ,QR1-502 - Abstract
Infection with highly pathogenic avian H5N1 influenza virus in humans often leads to severe respiratory disease with high mortality. Experimental infection in non-human primates can provide additional insight into disease pathogenesis. However, such a model should recapitulate the disease symptoms observed in humans, such as pneumonia and inflammatory cytokine response. While previous studies in macaques have demonstrated the occurrence of typical lesions in the lungs early after infection and a high level of immune activation, progression to severe disease and lethality were rarely observed. Here, we evaluated a routinely used combined route of infection via intra-bronchial, oral, and intra-nasal virus inoculation with aerosolized H5N1 exposure, with or without the regular collection of bronchoalveolar lavages early after infection. Both combined route and aerosol exposure resulted in similar levels of virus replication in nose and throat and similar levels of immune activation, cytokine, and chemokine release in the blood. However, while animals exposed to H5N1 by combined-route inoculation developed severe disease with high lethality, aerosolized exposure resulted in less lesions, as measured by consecutive computed tomography and less fever and lethal disease. In conclusion, not virus levels or immune activation, but route of infection determines fatal outcome for highly pathogenic avian H5N1 influenza infection.
- Published
- 2021
- Full Text
- View/download PDF
12. Retrospective Serology Study of Respiratory Virus Infections in Captive Great Apes
- Author
-
Hester Buitendijk, Zahra Fagrouch, Henk Niphuis, Willy M. Bogers, Kristin S. Warren, and Ernst J. Verschoor
- Subjects
respiratory viruses ,great apes ,serology ,anthroponotic infections ,Microbiology ,QR1-502 - Abstract
Great apes are extremely sensitive to infections with human respiratory viruses. In this study, we retrospectively analyzed sera from captive chimpanzees, gorillas and orang-utans. More than 1000 sera (403 chimpanzee, 77 gorilla, and 535 orang-utan sera) were analyzed for antibodies to the human respiratory viruses RSV (respiratory syncytial virus, hMPV (human metapneumovirus), H1N1 and H3N2 influenza A viruses, and influenza B virus. In all ape species high seroprevalences were found for RSV, hMPV, and influenza B virus. A high percentage of captive chimpanzees also showed evidence of influenza A H1N1 infections, and had low levels of H3N2 antibodies, while in sera from gorillas and orang-utans antibody levels to influenza A and B viruses were much lower or practically absent. Transmission of respiratory viruses was examined in longitudinal sera of young chimpanzees, and in chimpanzee sera taken during health checks. In young animals isolated cases of influenza infections were monitored, but evidence was found for single introductions followed by a rapid dissemination of RSV and hMPV within the group. Implementation of strict guidelines for handling and housing of nonhuman primates was shown to be an efficient method to reduce the introduction of respiratory infections in colonies of captive animals. RSV seroprevalence rates of chimpanzees remained high, probably due to circulating virus in the chimpanzee colony.
- Published
- 2014
- Full Text
- View/download PDF
13. Pandemic Swine-Origin H1N1 Influenza Virus Replicates to Higher Levels and Induces More Fever and Acute Inflammatory Cytokines in Cynomolgus versus Rhesus Monkeys and Can Replicate in Common Marmosets.
- Author
-
Petra Mooij, Gerrit Koopman, Daniëlla Mortier, Melanie van Heteren, Herman Oostermeijer, Zahra Fagrouch, Rudy de Laat, Gary Kobinger, Yan Li, Edmond J Remarque, Ivanela Kondova, Ernst J Verschoor, and Willy M J M Bogers
- Subjects
Medicine ,Science - Abstract
The close immunological and physiological resemblance with humans makes non-human primates a valuable model for studying influenza virus pathogenesis and immunity and vaccine efficacy against infection. Although both cynomolgus and rhesus macaques are frequently used in influenza virus research, a direct comparison of susceptibility to infection and disease has not yet been performed. In the current study a head-to-head comparison was made between these species, by using a recently described swine-origin pandemic H1N1 strain, A/Mexico/InDRE4487/2009. In comparison to rhesus macaques, cynomolgus macaques developed significantly higher levels of virus replication in the upper airways and in the lungs, involving both peak level and duration of virus production, as well as higher increases in body temperature. In contrast, clinical symptoms, including respiratory distress, were more easily observed in rhesus macaques. Expression of sialyl-α-2,6-Gal saccharides, the main receptor for human influenza A viruses, was 50 to 73 times more abundant in trachea and bronchus of cynomolgus macaques relative to rhesus macaques. The study also shows that common marmosets, a New World non-human primate species, are susceptible to infection with pandemic H1N1. The study results favor the cynomolgus macaque as model for pandemic H1N1 influenza virus research because of the more uniform and high levels of virus replication, as well as temperature increases, which may be due to a more abundant expression of the main human influenza virus receptor in the trachea and bronchi.
- Published
- 2015
- Full Text
- View/download PDF
14. Experimental infection of rhesus macaques and common marmosets with a European strain of West Nile virus.
- Author
-
Babs E Verstrepen, Zahra Fagrouch, Melanie van Heteren, Hester Buitendijk, Tom Haaksma, Niels Beenhakker, Giorgio Palù, Justin M Richner, Michael S Diamond, Willy M Bogers, Luisa Barzon, Stefan Chabierski, Sebastian Ulbert, Ivanela Kondova, and Ernst J Verschoor
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
West Nile virus (WNV) is a mosquito-borne flavivirus that infects humans and other mammals. In some cases WNV causes severe neurological disease. During recent years, outbreaks of WNV are increasing in worldwide distribution and novel genetic variants of the virus have been detected. Although a substantial amount of data exists on WNV infections in rodent models, little is known about early events during WNV infection in primates, including humans. To gain a deeper understanding of this process, we performed experimental infections of rhesus macaques and common marmosets with a virulent European WNV strain (WNV-Ita09) and monitored virological, hematological, and biochemical parameters. WNV-Ita09 productively infected both monkey species, with higher replication and wider tissue distribution in common marmosets compared to rhesus macaques. The animals in this study however, did not develop clinical signs of WNV disease, nor showed substantial deviations in clinical laboratory parameters. In both species, the virus induced a rapid CD56dimCD16bright natural killer response, followed by IgM and IgG antibody responses. The results of this study show that healthy rhesus macaques and common marmosets are promising animal models to study WNV-Ita09 infection. Both models may be particularly of use to evaluate potential vaccine candidates or to investigate WNV pathogenesis.
- Published
- 2014
- Full Text
- View/download PDF
15. Vaccine-induced protection of rhesus macaques against plasma viremia after intradermal infection with a European lineage 1 strain of West Nile virus.
- Author
-
Babs E Verstrepen, Herman Oostermeijer, Zahra Fagrouch, Melanie van Heteren, Henk Niphuis, Tom Haaksma, Ivanela Kondova, Willy M Bogers, Marina de Filette, Niek Sanders, Linda Stertman, Sofia Magnusson, Orsolya Lőrincz, Julianna Lisziewicz, Luisa Barzon, Giorgio Palù, Michael S Diamond, Stefan Chabierski, Sebastian Ulbert, and Ernst J Verschoor
- Subjects
Medicine ,Science - Abstract
The mosquito-borne West Nile virus (WNV) causes human and animal disease with outbreaks in several parts of the world including North America, the Mediterranean countries, Central and East Europe, the Middle East, and Africa. Particularly in elderly people and individuals with an impaired immune system, infection with WNV can progress into a serious neuroinvasive disease. Currently, no treatment or vaccine is available to protect humans against infection or disease. The goal of this study was to develop a WNV-vaccine that is safe to use in these high-risk human target populations. We performed a vaccine efficacy study in non-human primates using the contemporary, pathogenic European WNV genotype 1a challenge strain, WNV-Ita09. Two vaccine strategies were evaluated in rhesus macaques (Macaca mulatta) using recombinant soluble WNV envelope (E) ectodomain adjuvanted with Matrix-M, either with or without DNA priming. The DNA priming immunization was performed with WNV-DermaVir nanoparticles. Both vaccination strategies successfully induced humoral and cellular immune responses that completely protected the macaques against the development of viremia. In addition, the vaccine was well tolerated by all animals. Overall, The WNV E protein adjuvanted with Matrix-M is a promising vaccine candidate for a non-infectious WNV vaccine for use in humans, including at-risk populations.
- Published
- 2014
- Full Text
- View/download PDF
16. In vitro neutralization of low dose inocula at physiological concentrations of a monoclonal antibody which protects macaques against SHIV challenge.
- Author
-
David Davis, Wim Koornstra, Zahra Fagrouch, Ernst J Verschoor, Jonathan L Heeney, and Willy M J M Bogers
- Subjects
Medicine ,Science - Abstract
BACKGROUND: Passive transfer of antibodies can be protective in the simian human immunodeficiency virus (SHIV)--rhesus macaque challenge model. The human monoclonal antibody IgG1 b12 neutralizes human immunodeficiency type 1 (HIV-1) in vitro and protects against challenge by SHIV. Our hypothesis is that neutralizing antibodies can only completely inactivate a relatively small number of infectious virus. METHODS AND FINDINGS: We have used GHOST cell assays to quantify individual infectious events with HIV-1SF162 and its SHIV derivatives: the relatively neutralization sensitive SHIV(SF162P4) isolate and the more resistant SHIV(SF162P3). A plot of the number of fluorescent GHOST cells with increasing HIV-1SF162 dose is not linear. It is likely that with high-dose inocula, infection with multiple virus produces additive fluorescence in individual cells. In studies of the neutralization kinetics of IgG1 b12 against these isolates, events during the absorption phase of the assay, as well as the incubation phase, determine the level of neutralization. It is possible that complete inactivation of a virus is limited to the time it is exposed on the cell surface. Assays can be modified so that neutralization of these very low doses of virus can be quantified. A higher concentration of antibody is required to neutralize the same dose of resistant SHIV(SF162P3) than the sensitive SHIV(SF162P4). In the absence of selection during passage, the density of the CCR5 co-receptor on the GHOST cell surface is reduced. Changes in the CD4 : CCR5 density ratio influence neutralization. CONCLUSIONS: Low concentrations of IgG1 b12 completely inactivate small doses of the neutralization resistant SHIV(SF162P3). Assays need to be modified to quantify this effect. Results from modified assays may predict protection following repeated low-dose shiv challenges in rhesus macaques. It should be possible to induce this level of antibody by vaccination so that modified assays could predict the outcome of human trials.
- Published
- 2013
- Full Text
- View/download PDF
17. Protection in macaques immunized with HIV-1 candidate vaccines can be predicted using the kinetics of their neutralizing antibodies.
- Author
-
David Davis, Wim Koornstra, Daniella Mortier, Zahra Fagrouch, Ernst J Verschoor, Jonathan L Heeney, and Willy M J M Bogers
- Subjects
Medicine ,Science - Abstract
A vaccine is needed to control the spread of human immunodeficiency virus type 1 (HIV-1). An in vitro assay that can predict the protection induced by a vaccine would facilitate the development of such a vaccine. A potential candidate would be an assay to quantify neutralization of HIV-1.We have used sera from rhesus macaques that have been immunized with HIV candidate vaccines and subsequently challenged with simian human immunodeficiency virus (SHIV). We compared neutralization assays with different formats. In experiments with the standardized and validated TZMbl assay, neutralizing antibody titers against homologous SHIV(SF162P4) pseudovirus gave a variable correlation with reductions in plasma viremia levels. The target cells used in the assays are not just passive indicators of virus infection but are actively involved in the neutralization process. When replicating virus was used with GHOST cell assays, events during the absorption phase, as well as the incubation phase, determine the level of neutralization. Sera that are associated with protection have properties that are closest to the traditional concept of neutralization: the concentration of antibody present during the absorption phase has no effect on the inactivation rate. In GHOST assays, events during the absorption phase may inactivate a fixed number, rather than a proportion, of virus so that while complete neutralization can be obtained, it can only be found at low doses particularly with isolates that are relatively resistant to neutralization.Two scenarios have the potential to predict protection by neutralizing antibodies at concentrations that can be induced by vaccination: antibodies that have properties close to the traditional concept of neutralization may protect against a range of challenge doses of neutralization sensitive HIV isolates; a window of opportunity also exists for protection against isolates that are more resistant to neutralization but only at low challenge doses.
- Published
- 2011
- Full Text
- View/download PDF
18. Blocking NS3–NS4B interaction inhibits dengue virus in non-human primates
- Author
-
Olivia Goethals, Suzanne J. F. Kaptein, Bart Kesteleyn, Jean-François Bonfanti, Liesbeth Van Wesenbeeck, Dorothée Bardiot, Ernst J. Verschoor, Babs E. Verstrepen, Zahra Fagrouch, J. Robert Putnak, Dominik Kiemel, Oliver Ackaert, Roel Straetemans, Sophie Lachau-Durand, Peggy Geluykens, Marjolein Crabbe, Kim Thys, Bart Stoops, Oliver Lenz, Lotke Tambuyzer, Sandra De Meyer, Kai Dallmeier, Michael K. McCracken, Gregory D. Gromowski, Wiriya Rutvisuttinunt, Richard G. Jarman, Nicos Karasavvas, Franck Touret, Gilles Querat, Xavier de Lamballerie, Laurent Chatel-Chaix, Gregg N. Milligan, David W. C. Beasley, Nigel Bourne, Alan D. T. Barrett, Arnaud Marchand, Tim H. M. Jonckers, Pierre Raboisson, Kenny Simmen, Patrick Chaltin, Ralf Bartenschlager, Willy M. Bogers, Johan Neyts, and Marnix Van Loock
- Subjects
Multidisciplinary - Abstract
Dengue is a major health threat and the number of symptomatic infections caused by the four dengue serotypes is estimated to be 96 million1 with annually around 10,000 deaths2. However, no antiviral drugs are available for the treatment or prophylaxis of dengue. We recently described the interaction between non-structural proteins NS3 and NS4B as a promising target for the development of pan-serotype dengue virus (DENV) inhibitors3. Here we present JNJ-1802—a highly potent DENV inhibitor that blocks the NS3–NS4B interaction within the viral replication complex. JNJ-1802 exerts picomolar to low nanomolar in vitro antiviral activity, a high barrier to resistance and potent in vivo efficacy in mice against infection with any of the four DENV serotypes. Finally, we demonstrate that the small-molecule inhibitor JNJ-1802 is highly effective against viral infection with DENV-1 or DENV-2 in non-human primates. JNJ-1802 has successfully completed a phase I first-in-human clinical study in healthy volunteers and was found to be safe and well tolerated4. These findings support the further clinical development of JNJ-1802, a first-in-class antiviral agent against dengue, which is now progressing in clinical studies for the prevention and treatment of dengue.
- Published
- 2023
- Full Text
- View/download PDF
19. Microbial molecule ingress promotes neuroinflammation and brain CCR5 expression in persons with HIV-associated neurocognitive disorders
- Author
-
William G, Branton, Jason P, Fernandes, Nazanin, Mohammadzadeh, Mathew A L, Doan, Jon D, Laman, Benjamin B, Gelman, Zahra, Fagrouch, Ivanela, Kondova, Petra, Mooij, Gerrit, Koopman, Christopher, Power, and Translational Immunology Groningen (TRIGR)
- Subjects
Receptors, CCR5 ,Endocrine and Autonomic Systems ,Immunology ,Neurocognitive Disorders ,HIV ,Brain ,HIV Infections ,Peptidoglycan ,Behavioral Neuroscience ,Neuroinflammation ,SIV ,Glia ,Neuroinflammatory Diseases ,HIV-1 ,Humans ,CCR5 - Abstract
Background: Systemic inflammation accompanies HIV-1 infection, resulting in microbial translocation from different tissues. We investigated interactions between lentivirus infections, neuroinflammation and microbial molecule presence in the brain. Methods: Brain tissues from adult humans with (n = 22) and without HIV-1 (n = 11) infection as well as adult nonhuman primates (NHPs) with (n = 11) and without (n = 4) SIVmac251 infection were investigated by RT-PCR/ddPCR, immunofluorescence and western blotting. Studies of viral infectivity, host immune gene expression and viability were performed in primary human neural cells. Findings: Among NHPs, SIV DNA quantitation in brain showed increased levels among animals with SIV encephalitis (n = 5) that was associated with bacterial genomic copy number as well as CCR5 and CASP1 expression in brain. Microbial DnaK and peptidoglycan were immunodetected in brains from uninfected and SIV-infected animals, chiefly in glial cells. Human microglia infected by HIV-1 showed increased p24 production after exposure to peptidoglycan that was associated CCR5 induction. HIV-1 Vpr application to human neurons followed by peptidoglycan exposure resulted in reduced mitochondrial function and diminished beta-III tubulin expression. In human brains, bacterial genome copies (250–550 copies/gm of tissue), were correlated with increased bacterial rRNA and GroEL transcript levels in patients with HIV-associated neurocognitive disorders (HAND). Glial cells displayed microbial GroEL and peptidoglycan immunoreactivity accompanied by CCR5 induction in brains from patients with HAND. Interpretation: Increased microbial genomes and proteins were evident in brain tissues from lentivirus-infected humans and animals and associated with neurological disease. Microbial molecule translocation into the brain might exacerbate neuroinflammatory disease severity and represent a driver of lentivirus-associated brain disease.
- Published
- 2023
20. Novel application of [
- Author
-
Lisette, Meijer, Kinga P, Böszörményi, Jaco, Bakker, Gerrit, Koopman, Petra, Mooij, Dagmar, Verel, Zahra, Fagrouch, Babs E, Verstrepen, Uta, Funke, Martien P J, Mooijer, Jan A M, Langermans, Ernst J, Verschoor, Albert D, Windhorst, and Marieke A, Stammes
- Subjects
Pyrimidines ,SARS-CoV-2 ,Positron Emission Tomography Computed Tomography ,Animals ,COVID-19 ,Pyrazoles ,Pneumonia ,Lung ,Macaca mulatta - Abstract
The aim of this study was to investigate the application of [Four experimentally SARS-CoV-2 infected rhesus monkeys were followed for seven weeks post infection (pi) with a weekly PET-CT using [All animals were positive for SARS-CoV-2 in both the swabs and BALs on multiple timepoints pi. The initial development of pulmonary lesions was already detected at the first scan, performed 2-days pi. PET revealed an increased tracer uptake in the pulmonary lesions and mediastinal lymph nodes of all animals from the first scan obtained after infection and onwards. However, also an increased uptake was detected in the lung tissue surrounding the lesions, which persisted until day 30 and then subsided by day 37-44 pi. In parallel, a similar pattern of increased expression of activation markers was observed on dendritic cells in blood.This study illustrates that [
- Published
- 2022
21. Safety and Immunogenicity of Four-Segmented Rift Valley Fever Virus in the Common Marmoset
- Author
-
Paul J. Wichgers Schreur, Petra Mooij, Gerrit Koopman, Babs E. Verstrepen, Zahra Fagrouch, Daniella Mortier, Nikki van Driel, Jet Kant, Sandra van de Water, Willy M. Bogers, Carine Punt, Lucien van Keulen, Ernst J. Verschoor, and Jeroen Kortekaas
- Subjects
Host Pathogen Interaction & Diagnostics ,Pharmacology ,Bacteriologie ,Immunology ,Bacteriology ,Bacteriology, Host Pathogen Interaction & Diagnostics ,PE&RC ,Host Pathogen Interactie & Diagnostiek ,Virology & Molecular Biology ,Virologie & Moleculaire Biologie ,Infectious Diseases ,Bacteriologie, Host Pathogen Interactie & Diagnostiek ,Life Science ,Pharmacology (medical) - Abstract
Rift Valley fever virus (RVFV) is an emerging mosquito-borne bunyavirus that is highly pathogenic to wild- and domesticated ruminants, camelids and humans. While animals are exclusively infected via mosquito bites, humans can also be infected via contact with tissues or blood released during the slaughtering of RVFV-infected animals. No human vaccine is available and currently commercialized veterinary vaccines do not optimally combine efficacy with safety. We previously reported the development of two novel live-attenuated RVF vaccines, created by splitting the M genome segment and deleting the major virulence determinant NSs. The vaccine candidates, referred to as the veterinary vaccine vRVFV-4s and the human vaccine hRVFV-4s, were shown to induce protective immunity in multiple species after a single vaccination. Anticipating on accidental exposure of humans to the veterinary vaccine, and to evaluate the safety of the hRVFV-4s candidate vaccine for humans, the safety of each vaccine was evaluated in the most susceptible nonhuman primate model, the common marmoset (Callithrix jacchus). Marmosets were inoculated with high doses of each vaccine and were monitored for clinical signs as well as for vaccine virus dissemination, shedding and spreading to the environment. To accurately assess the attenuation of both vaccine viruses, separate groups of marmosets were inoculated with the parent wild-type RVFV strains. Both wild-type strains induced high viremia and disseminated to primary target organs, associated with mild- to severe morbidity, while both vaccines were well tolerated with absence of dissemination and shedding, while inducing potent neutralizing antibody responses. The results of the studies support the unprecedented safety profile of both vaccines for animals and humans.
- Published
- 2021
- Full Text
- View/download PDF
22. Influenza virus infection as well as immunization with DNA encoding haemagglutinin protein induces potent antibody-dependent phagocytosis (ADP) and monocyte infection-enhancing responses in macaques
- Author
-
Gerrit Koopman, Daniella Mortier, Petra Mooij, Ernst J. Verschoor, Edmond J. Remarque, Willy M. J. M. Bogers, Zahra Fagrouch, Samira Michels, and Sam O. Hofman
- Subjects
0301 basic medicine ,Neutrophils ,Phagocytosis ,030106 microbiology ,Fc receptor ,Heterologous ,Hemagglutinin Glycoproteins, Influenza Virus ,Antibodies, Viral ,Monocytes ,Virus ,03 medical and health sciences ,Orthomyxoviridae Infections ,Virology ,Vaccines, DNA ,medicine ,Animals ,Humans ,Cells, Cultured ,Antibody-dependent cell-mediated cytotoxicity ,biology ,Monocyte ,Dendritic Cells ,Orthomyxoviridae ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Immunization ,Influenza Vaccines ,biology.protein ,Macaca ,Antibody - Abstract
Antibodies directed against the conserved regions within the influenza A haemagglutinin (HA) protein are detected at low frequency in humans. These antibodies display a broad reactivity against divergent influenza virus strains and could potentially offer broad protection. The in vivo protective effect of these antibodies is mainly mediated through Fc receptor effector functions. While antibody-dependent phagocytosis (ADP) of anti-HA antibodies has been demonstrated in human sera and sera from influenza virus-infected macaques, it is not known whether ADP can also be induced by vaccination and what the relative strength of ADP responses is in comparison to other antibody functions. Using a cohort of influenza virus-infected and immunized macaques, we demonstrate that infection as well as immunization with DNA-encoding HA induces high-titre ADP responses against HA, which are of potency 100-1000 times higher than virus inhibitory functions including antibody-dependent cell-mediated cytotoxicity (ADCC), virus neutralization (VN) and haemagglutinin inhibition (HAI). ADP activity was equally high against HA of heterologous influenza strains of the same subtype, in contrast to virus inhibitory functions, which were all greatly diminished. ADP titres against H5, representing a hetero-subtypic virus, were much lower. ADP was measured in THP-1 cells but was also observed in primary peripheral blood monocytes and neutrophils. Furthermore, at high serum dilution enhanced infection of both monocytes and myeloid dendritic cells (mDC) was observed. Hence, influenza virus infection as well as DNA-immunization against HA can induce high-titre ADP responses that can potentially enhance influenza virus infection of primary phagocytic and dendritic cells.
- Published
- 2019
- Full Text
- View/download PDF
23. Bronchoalveolar lavage affects thorax computed tomography of healthy and SARS-CoV-2 infected rhesus macaques (Macaca mulatta)
- Author
-
Dian G M Zijlmans, Jaco Bakker, Leo van Geest, Jan A.M. Langermans, Lisette Meijer, Zahra Fagrouch, Annemiek Maaskant, Babs E. Verstrepen, Ernst J. Verschoor, Marieke A. Stammes, and AISS LAS/3'R Centre ULS
- Subjects
Thorax ,RNA viruses ,Pathology ,Viral Diseases ,Pulmonology ,Coronaviruses ,Computed tomography ,Monkeys ,Diagnostic Radiology ,0403 veterinary science ,Medical Conditions ,Medicine ,Lung ,Tomography ,Optical Properties ,Pathology and laboratory medicine ,Mammals ,0303 health sciences ,Multidisciplinary ,medicine.diagnostic_test ,Radiology and Imaging ,Eukaryota ,04 agricultural and veterinary sciences ,Animal Models ,Medical microbiology ,respiratory system ,medicine.anatomical_structure ,Infectious Diseases ,Experimental Organism Systems ,Vertebrates ,Viruses ,Physical Sciences ,SARS CoV 2 ,Pathogens ,Anatomy ,Bronchoalveolar Lavage Fluid ,Macaque ,Research Article ,Primates ,Opacity ,medicine.medical_specialty ,SARS coronavirus ,040301 veterinary sciences ,Imaging Techniques ,Science ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Materials Science ,Material Properties ,Neuroimaging ,Research and Analysis Methods ,Microbiology ,Diagnostic modalities ,03 medical and health sciences ,Respiratory Disorders ,Diagnostic Medicine ,Hounsfield scale ,Old World monkeys ,Animals ,General ,030304 developmental biology ,Medicine and health sciences ,Biology and life sciences ,Rhesus Monkeys ,business.industry ,SARS-CoV-2 ,Organisms ,Viral pathogens ,COVID-19 ,Covid 19 ,Macaca mulatta ,Microbial pathogens ,Computed Axial Tomography ,respiratory tract diseases ,Disease Models, Animal ,Bronchoalveolar lavage ,Amniotes ,Respiratory Infections ,Animal Studies ,business ,Tomography, X-Ray Computed ,Zoology ,Respiratory tract ,Neuroscience - Abstract
Medical imaging as method to assess the longitudinal process of a SARS-CoV-2 infection in non-human primates is commonly used in research settings. Bronchoalveolar lavage (BAL) is regularly used to determine the local virus production and immune effects of SARS-CoV-2 in the lower respiratory tract. However, the potential interference of those two diagnostic modalities is unknown in non-human primates. The current study investigated the effect and duration of BAL on computed tomography (CT) in both healthy and experimentally SARS-CoV-2-infected female rhesus macaques (Macaca mulatta). In addition, the effect of subsequent BALs was reviewed. Thorax CTs and BALs were obtained from four healthy animals and 11 experimentally SARS-CoV-2-infected animals. From all animals, CTs were obtained just before BAL, and 24 hours post-BAL. Additionally, from the healthy animals, CTs immediately after, and four hours post-BAL were obtained. Thorax CTs were evaluated for alterations in lung density, measured in Hounsfield units, and a visual semi-quantitative scoring system. An increase in the lung density was observed on the immediately post-BAL CT but resolved within 24 hours in the healthy animals. In the infected animals, a significant difference in both the lung density and CT score was still found 24 hours after BAL. Furthermore, the differences between time points in CT score were increased for the second BAL. These results indicate that the effect of BAL on infected lungs is not resolved within the first 24 hours. Therefore, it is important to acknowledge the interference between BAL and CT in rhesus macaques.
- Published
- 2021
24. SARS-CoV-2 causes brain inflammation and induces Lewy body formation in macaques
- Author
-
Eva Roos, Babs E. Verstrepen, Ernst J. Verschoor, Marianna Bugiani, Lioe-Fee de Geus-Oei, Jan A.M. Langermans, Jacqueline Wubben, Ronald E. Bontrop, Willy M. J. M. Bogers, Diana Lozovagia, Zahra Fagrouch, Bernadette Schurink, Amber Q. Mayenburg, Nikki van Driel, Marieke A. Stammes, Ingrid H.C.H.M. Philippens, Jinte Middeldorp, and Kinga P. Böszörményi
- Subjects
Pituitary gland ,Pathology ,medicine.medical_specialty ,Lewy body ,biology ,Microglia ,business.industry ,Inflammation ,Neuropathology ,medicine.disease ,Asymptomatic ,Macaque ,Virus ,medicine.anatomical_structure ,biology.animal ,medicine ,medicine.symptom ,business - Abstract
SARS-CoV-2 may cause acute respiratory disease, but the infection can also initiate neurological symptoms. Here we show that SARS-CoV-2 infection causes brain inflammation in the macaque model. An increased metabolic activity in the pituitary gland of two macaques was observed by longitudinal positron emission tomography-computed tomography (PET-CT). Post-mortem analysis demonstrated infiltration of T-cells and activated microglia in the brain, and viral RNA was detected in brain tissues from one animal. We observed Lewy bodies in brains of all rhesus macaques. These data emphasize the virus’ capability to induce neuropathology in this nonhuman primate model for SARS-CoV-2 infection. As in humans, Lewy body formation is an indication for the development of Parkinson’s disease, this data represents a warning for potential long-term neurological effects after SARS-CoV-2 infection.TeaserSARS-CoV-2 causes brain inflammation and Lewy bodies, a hallmark for Parkinson, after an asymptomatic infection in macaques.
- Published
- 2021
- Full Text
- View/download PDF
25. A Bacterially-Expressed Recombinant Envelope Protein from Usutu Virus Induces Neutralizing Antibodies in Rabbits
- Author
-
Montserrat Bárcena, Ernst J. Verschoor, Amy van Dijke, Babs E. Verstrepen, Zahra Fagrouch, Willy M. J. M. Bogers, Gesine Cornellissen, Janet Hirsch, Soesjiel Dwarka, Ronald W. A. L. Limpens, Boyd Aaldijk, Bart W. Faber, Kinga P. Böszörményi, Gwendoline Kiemenyi Kayere, Roberto Rodriguez Garcia, Bram Koster, Nicole Heijmans, and Clemens H. M. Kocken
- Subjects
0301 basic medicine ,Protein subunit ,viruses ,030231 tropical medicine ,Immunology ,lcsh:Medicine ,Article ,law.invention ,Zika virus ,03 medical and health sciences ,0302 clinical medicine ,flavivirus ,law ,Drug Discovery ,Pharmacology (medical) ,emerging disease ,neutralizing antibodies ,Usutu virus ,Pharmacology ,recombinant E protein ,biology ,lcsh:R ,biology.organism_classification ,Virology ,In vitro ,3. Good health ,Flavivirus ,USUV ,030104 developmental biology ,Infectious Diseases ,Immunization ,vaccine development ,biology.protein ,Recombinant DNA ,Antibody - Abstract
Background: Recently, an emerging flavivirus, Usutu virus (USUV), has caused an epidemic among birds in Europe, resulting in a massive die-off in Eurasian blackbirds. Currently found only in Europe and Africa, it can be envisioned that Usutu virus will follow the path of other flaviviruses, like West Nile virus and Zika virus, and will spread via its mosquito vectors and bird hosts to other parts of the world. Several cases of human infections by Usutu virus have already been published. Anticipating this spread, development of an efficacious vaccine would be highly desirable. Method: This study describes the production in E. coli, purification, and refolding of a partial USUV envelope protein. Prior to immunization, the protein was characterized using size exclusion chromatography, transmission electron microscopy and dynamic light scattering, showing the limited presence of virus-like structures, indicating that the protein solution is probably a mixture of mono and multimeric envelope proteins. Results: Immunizations of two rabbits with the refolded E-protein fraction, mixed with a strong adjuvant, resulted in the generation of neutralizing antibodies, as evidenced in an in vitro assay. Discussion: The way forward towards a subunit vaccine against Usutu virus infection is discussed.
- Published
- 2021
26. The post-acute phase of SARS-CoV-2 infection in two macaques species is associated with signs of ongoing virus replication and pathology in pulmonary and extrapulmonary tissues
- Author
-
Ronald E. Bontrop, Herman Oostermeijer, E. J. Remarque, Ernst J. Verschoor, Ella A. Zuiderwijk-Sick, Gerrit Koopman, Petra Mooij, Roja Fidel Acar, Nikki van Driel, Deborah Adema, Boudewijn Ouwerling, Patricia Sastre, Ivonne G. Nieuwenhuis, Daniella Mortier, Babs E. Verstrepen, Bart L. Haagmans, Alexis C. R. Hoste, Tom Haaksma, Willy M. J. M. Bogers, Gwendoline Kiemenyi-Kayere, Zahra Fagrouch, Ivanela Kondova, Lisette Meijer, Marieke A. Stammes, Jan A.M. Langermans, Richard A. W. Vervenne, Kinga P. Böszörményi, and Henk Niphuis
- Subjects
medicine.medical_specialty ,Pathology ,Conjunctiva ,business.industry ,Tracheobronchial lymph nodes ,Virus ,medicine.anatomical_structure ,Viral replication ,medicine ,Mesenteric lymph nodes ,Histopathology ,Lymph ,business ,Respiratory tract - Abstract
The post-acute phase of SARS-CoV-2 infection was investigated in rhesus macaques (Macaca mulatta) and cynomolgus macaques (Macaca fascicularis). During the acute phase of infection, SARS-CoV-2 was shed via nose and throat, and viral RNA was occasionally detected in feces. This phase coincided with a transient change in systemic immune activation. Even after the alleged resolution of the infection, as suggested by the absence of viral RNA in nasal and tracheal swabs, computed tomography (CT) and positron emission tomography (PET)-CT were able to reveal pulmonary lesions and activated tracheobronchial lymph nodes in all animals. Post-mortem histological examination of the lung tissue revealed mostly marginal or resolving minimal lesions that were indicative of SARS-CoV-2 infection. Evidence for SARS-CoV-2-induced histopathology was also found in extrapulmonary tissue samples, like conjunctiva, cervical and mesenteric lymph nodes.However, 5-6 weeks after SARS-CoV-2 exposure, upon necropsy, viral RNA was still detectable in a wide range of tissue samples in 50% of the macaques and included amongst others the heart, the respiratory tract and surrounding lymph nodes, salivary gland, and conjunctiva. Subgenomic messenger RNA was detected in the lungs and tracheobronchial lymph nodes, indicative of ongoing virus replication during the post-acute phase. These results could be relevant for understanding the long-term consequences of COVID-19 in humans.Author summaryMore than a year after the start of the pandemic, the long-term consequences of SARS-CoV-2 infection start to surface. The variety of clinical manifestations associated with post-acute COVID-19 suggests the involvement of multiple biological mechanisms. In this study, we show that rhesus and cynomolgus macaques shed virus from their respiratory tract, generate virus-specific humoral immune responses, and show signs of SARS-CoV-2-induced lung pathology. PET-CT revealed that both species showed ongoing mild to moderate pulmonary disease, even after the virus was no longer detectable in nasal and tracheal swabs. Five to six weeks after infection, necropsy confirmed minimal to mild histopathological manifestations in various tissues, like the lungs, heart, lymph nodes, and conjunctiva. We detected Viral RNA in the heart, respiratory tract, and tracheobronchial lymph nodes, and subgenomic messenger RNA in the lungs and surrounding lymph nodes, indicative of ongoing virus replication. We show widespread tissue dissemination of SARS-CoV-2 in infected macaques and the presence of replicating virus in lungs and surrounding lymph nodes after alleged convalescence of infection. This finding is intriguing in the light of long-COVID disease symptoms seen in humans as it has been hypothesized that persistent infection may contribute to this phenomenon.
- Published
- 2020
- Full Text
- View/download PDF
27. Aerosolized pH1N1 influenza infection induces less systemic and local immune activation in the lung than combined intrabronchial, nasal and oral exposure in cynomolgus macaques
- Author
-
Petra Mooij, Daniella Mortier, Marieke A. Stammes, Willy M. J. M. Bogers, Ernst J. Verschoor, Zahra Fagrouch, and Gerrit Koopman
- Subjects
0301 basic medicine ,Male ,Chemokine ,Inflammation ,Bronchi ,Biology ,Nose ,Virus Replication ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Influenza A Virus, H1N1 Subtype ,Orthomyxoviridae Infections ,Virology ,Lymphopenia ,Influenza, Human ,medicine ,Animals ,Humans ,030212 general & internal medicine ,Lung ,Immunity, Cellular ,Mouth ,Monocyte ,Immunogenicity ,respiratory system ,Immunity, Humoral ,Virus Shedding ,Disease Models, Animal ,Macaca fascicularis ,030104 developmental biology ,medicine.anatomical_structure ,Viral replication ,Immunology ,biology.protein ,Cytokines ,medicine.symptom ,Bronchoalveolar Lavage Fluid - Abstract
Non-human primates form an important animal model for the evaluation of immunogenicity and efficacy of novel ‘universal’ vaccine candidates against influenza virus. However, in most studies a combination of intra-tracheal or intra-bronchial, oral and nasal virus inoculation is used with a standard virus dose of between 1 and 10 million tissue culture infective doses, which differs from typical modes of virus exposure in humans. This paper studies the systemic and local inflammatory and immune effects of aerosolized versus combined-route exposure to pandemic H1N1 influenza virus. In agreement with a previous study, both combined-route and aerosol exposure resulted in similar levels of virus replication in nose, throat and lung lavages. However, the acute release of pro-inflammatory cytokines and chemokines, acute monocyte activation in peripheral blood as well as increased cytokine production and T-cell proliferation in the lungs were only observed after combined-route infection and not after aerosol exposure. Longitudinal evaluation by computed tomography demonstrated persistence of lung lesions after resolution of the infection and a tendency for more lesions in the lower lung lobes after combined-route exposure versus upper and middle lung lobes after aerosol exposure. Computed tomography scores were observed to correlate with fever. In conclusion, influenza virus infection by aerosol exposure is accompanied by less immune-activation and inflammation in comparison with direct virus installation, despite similar levels of virus replication and development of lesions in the lungs.
- Published
- 2020
28. Targeted Diet Modification Reduces Multiple Sclerosis-like Disease in Adult Marmoset Monkeys from an Outbred Colony
- Author
-
Jan Bauer, Bart J. L. Eggen, Bert A. 't Hart, Corien Grit, Marissa L. Dubbelaar, Ernst J. Verschoor, Nikki van Driel, Carien Bus-Spoor, Yolanda S. Kap, Jon D. Laman, Hermie J. M. Harmsen, Zahra Fagrouch, Susanne M. Kooistra, Molecular Neuroscience and Ageing Research (MOLAR), Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI), Microbes in Health and Disease (MHD), Translational Immunology Groningen (TRIGR), and Restoring Organ Function by Means of Regenerative Medicine (REGENERATE)
- Subjects
0301 basic medicine ,Herpesvirus 3, Human ,Encephalomyelitis, Autoimmune, Experimental ,Multiple Sclerosis ,Diet therapy ,Cells ,Encephalomyelitis ,Immunology ,Apoptosis ,Gut flora ,Myelin oligodendrocyte glycoprotein ,03 medical and health sciences ,0302 clinical medicine ,RNA, Ribosomal, 16S ,biology.animal ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cells, Cultured ,biology ,Sequence Analysis, RNA ,Multiple sclerosis ,Experimental autoimmune encephalomyelitis ,Brain ,Marmoset ,Callithrix ,medicine.disease ,biology.organism_classification ,Gastrointestinal Microbiome ,Disease Models, Animal ,030104 developmental biology ,Spinal Cord ,Dietary Supplements ,biology.protein ,Myelin-Oligodendrocyte Glycoprotein ,Bifidobacterium ,030217 neurology & neurosurgery ,Demyelinating Diseases ,Diet Therapy - Abstract
Experimental autoimmune encephalomyelitis (EAE) in common marmosets is a translationally relevant model of the chronic neurologic disease multiple sclerosis. Following the introduction of a new dietary supplement in our purpose-bred marmoset colony, the percentage of marmosets in which clinically evident EAE could be induced by sensitization against recombinant human myelin oligodendrocyte glycoprotein in IFA decreased from 100 to 65%. The reduced EAE susceptibility after the dietary change coincided with reduced Callitrichine herpesvirus 3 expression in the colony, an EBV-related γ1-herpesvirus associated with EAE. We then investigated, in a controlled study in marmoset twins, which disease-relevant parameters were affected by the dietary change. The selected twins had been raised on the new diet for at least 12 mo prior to the study. In twin siblings reverted to the original diet 8 wk prior to EAE induction, 100% disease prevalence (eight out of eight) was restored, whereas in siblings remaining on the new diet the EAE prevalence was 75% (six out of eight). Spinal cord demyelination, a classical hallmark of the disease, was significantly lower in new-diet monkeys than in monkeys reverted to the original diet. In new-diet monkeys, the proinflammatory T cell response to recombinant human myelin oligodendrocyte glycoprotein was significantly reduced, and RNA-sequencing revealed reduced apoptosis and enhanced myelination in the brain. Systematic typing of the marmoset gut microbiota using 16S rRNA sequencing demonstrated a unique, Bifidobacteria-dominated composition, which changed after disease induction. In conclusion, targeted dietary intervention exerts positive effects on EAE-related parameters in multiple compartments of the marmoset’s gut-immune–CNS axis.
- Published
- 2018
- Full Text
- View/download PDF
29. Serological evidence of coronavirus infections in native hamadryas baboons (Papio hamadryas hamadryas) of the Kingdom of Saudi Arabia
- Author
-
Zahra Fagrouch, Ernst J. Verschoor, Khalid Al-Hezaimi, R V Herwijnen, A. O. Olarinmoye, Hester Buitendijk, A Boug, Olayinka Olabisi Ishola, Henk Niphuis, and Babasola O Olugasa
- Subjects
0301 basic medicine ,Male ,Epidemiology ,Middle East respiratory syndrome coronavirus ,viruses ,coronaviruses ,Kingdom of Saudi Arabia ,030106 microbiology ,Saudi Arabia ,Serological evidence ,Commensal ,medicine.disease_cause ,Hamadryas ,03 medical and health sciences ,Seroepidemiologic Studies ,biology.animal ,Zoonoses ,medicine ,Prevalence ,Animals ,infections ,Hamadryas baboon ,Papio hamadryas ,biology ,Monkey Diseases ,biology.organism_classification ,Virology ,Original Papers ,Coronavirus ,030104 developmental biology ,Infectious Diseases ,biology.protein ,Middle East Respiratory Syndrome Coronavirus ,Female ,Papio hamadryas hamadryas ,Antibody ,Coronavirus Infections ,Baboon - Abstract
SUMMARYThe hamadryas baboon (Papio hamadryas hamadryas) is the only indigenous species of non-human primates (NHP) found in the Kingdom of Saudi Arabia (KSA). There are no peer-reviewed publications on viral infections of the baboons of KSA. Apart from camels, other animals are likely sources of the novel Middle East Respiratory Syndrome coronavirus (MERSCoV) for humans. We investigated evidence of highly pathogenic coronavirus infections including MERSCoV in a large group of commensal baboons accompanied by feral dogs, on the outskirts of Ta'if city, KSA, in February 2013. Fifty baboons (16 juveniles and 34 adults) were screened for serum antibodies to human coronaviruses (HCoV-043/-NL63/-229) and canine coronaviruses (CCoV-1-3) using direct Enzyme-linked Immunosorbent Assay (ELISA) technique and for MERSCoV antibodies using Serum Neutralization Test (SNT). Of the 50 sampled baboons, 22% (n= 11) were seropositive to HCoVs, 10% (n= 5) were seropositive to CCoVs, while none had detectable MERSCoV antibodies. These findings bear potentially significant implications for public health, canine health and baboon conservation efforts, necessitating follow-up investigations and preventive measures at locations where baboons frequent human habitations, or are regarded as tourist attractions, in KSA.
- Published
- 2017
30. Absence of accessory genes in a divergent simian T-lymphotropic virus type 1 isolated from a bonnet macaque (Macaca radiata)
- Author
-
Martin Deijs, Philippe V. Afonso, Willy M. J. M. Bogers, Antoine Gessain, Henk Niphuis, Lia van der Hoek, Zahra Fagrouch, Ernst J. Verschoor, Medical Microbiology and Infection Prevention, AII - Infectious diseases, Epidémiologie et Physiopathologie des Virus Oncogènes (EPVO (UMR_3569 / U-Pasteur_3)), Institut Pasteur [Paris]-Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), University of Amsterdam [Amsterdam] (UvA), This study received funding from the 'Investissement d’Avenir' as part of a 'Laboratoire d’Excellence' (LabEx) French research program: Integrative Biology of Emerging Infectious Diseases (ANR10-LBX-62 IBEID). It was also funded by an internal BPRC institutional grant., ANR-10-LABX-62-IBEID,IBEID,Laboratoire d'Excellence 'Integrative Biology of Emerging Infectious Diseases'(2010), ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), and Institut Pasteur [Paris] (IP)-Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,viruses ,RC955-962 ,Artificial Gene Amplification and Extension ,Simian-T-lymphotropic virus ,Monkeys ,[SDV.BID.SPT]Life Sciences [q-bio]/Biodiversity/Systematics, Phylogenetics and taxonomy ,Polymerase Chain Reaction ,Macaque ,Database and Informatics Methods ,0302 clinical medicine ,Arctic medicine. Tropical medicine ,Viral Regulatory and Accessory Proteins ,Phylogeny ,ComputingMilieux_MISCELLANEOUS ,Data Management ,Mammals ,Genetics ,Deltaretrovirus Infections ,[SDV.BID.EVO]Life Sciences [q-bio]/Biodiversity/Populations and Evolution [q-bio.PE] ,Eukaryota ,Phylogenetic Analysis ,Genomics ,3. Good health ,Phylogenetics ,Infectious Diseases ,Vertebrates ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,Public aspects of medicine ,RA1-1270 ,Simian T-lymphotropic virus 1 ,Sequence Analysis ,Research Article ,Primates ,Computer and Information Sciences ,Bioinformatics ,Sequence analysis ,030231 tropical medicine ,India ,Biology ,Research and Analysis Methods ,Virus ,Open Reading Frames ,03 medical and health sciences ,Complete sequence ,Sequence Motif Analysis ,biology.animal ,Old World monkeys ,Animals ,Evolutionary Systematics ,Gene Prediction ,Molecular Biology Techniques ,Bonnet macaque ,Molecular Biology ,Gene ,Taxonomy ,Evolutionary Biology ,Biology and life sciences ,Organisms ,Public Health, Environmental and Occupational Health ,Computational Biology ,Sequence Analysis, DNA ,Genome Analysis ,biology.organism_classification ,Open reading frame ,Macaca radiata ,030104 developmental biology ,Amniotes ,Sequence Alignment - Abstract
Background Primate T-lymphotropic viruses type 1 (PTLV-1) are complex retroviruses infecting both human (HTLV-1) and simian (STLV-1) hosts. They share common epidemiological, clinical and molecular features. In addition to the canonical gag, pol, env retroviral genes, PTLV-1 purportedly encodes regulatory (i.e. Tax, Rex, and HBZ) and accessory proteins (i.e. P12/8, P13, P30). The latter have been found essential for viral persistence in vivo. Methodology/Principal findings We have isolated a STLV-1 virus from a bonnet macaque (Macaca radiata–Mra18C9), a monkey from India. The complete sequence was obtained and phylogenetic analyses were performed. The Mra18C9 strain is highly divergent from the known PTLV-1 strains. Intriguingly, the Mra18C9 lacks the 3 accessory open reading frames. In order to determine if the absence of accessory proteins is specific to this particular strain, a comprehensive analysis of the complete PTLV-1 genomes available in Genbank was performed and found that the lack of one or many accessory ORF is common among PTLV-1. Conclusion This study raises many questions regarding the actual nature, role and importance of accessory proteins in the PTLV-1 biology., Author summary Primate T-lymphotropic viruses type 1 (PTLV-1) are complex retroviruses infecting both human (HTLV-1) and simian (STLV-1) hosts. It has been shown that the persistence and pathogenesis of these viruses depend on the expression of small, accessory proteins. A bonnet macaque (a monkey present in India) was found infected with STLV-1. The genome was sequenced and found quite divergent from the other STLV-1 genomes previously described. Intriguingly, this virus does not encode accessory proteins. Analysis of other available sequences found that most strains lack at least one accessory gene. Thus the importance and the role of these proteins in the PTLV-1 biology should be revisited.
- Published
- 2019
- Full Text
- View/download PDF
31. Retrospective Serology Study of Respiratory Virus Infections in Captive Great Apes
- Author
-
Zahra Fagrouch, Ernst J. Verschoor, Hester Buitendijk, Willy M. J. M. Bogers, Kristin S. Warren, and Henk Niphuis
- Subjects
Pan troglodytes ,viruses ,lcsh:QR1-502 ,serology ,Gorilla ,Biology ,medicine.disease_cause ,Antibodies, Viral ,Virus ,lcsh:Microbiology ,Article ,Serology ,Human metapneumovirus ,Seroepidemiologic Studies ,Virology ,biology.animal ,respiratory viruses ,Influenza A virus ,medicine ,Seroprevalence ,Animals ,Respiratory Tract Infections ,Gorilla gorilla ,Respiratory tract infections ,Pongo ,Primate Diseases ,great apes ,anthroponotic infections ,virus diseases ,biology.organism_classification ,3. Good health ,respiratory tract diseases ,Respiratory Syncytial Viruses ,Influenza B virus ,Infectious Diseases ,Virus Diseases ,Immunology ,Respiratory virus ,Animals, Zoo ,Metapneumovirus - Abstract
Great apes are extremely sensitive to infections with human respiratory viruses. In this study, we retrospectively analyzed sera from captive chimpanzees, gorillas and orang-utans. More than 1000 sera (403 chimpanzee, 77 gorilla, and 535 orang-utan sera) were analyzed for antibodies to the human respiratory viruses RSV (respiratory syncytial virus, hMPV (human metapneumovirus), H1N1 and H3N2 influenza A viruses, and influenza B virus. In all ape species high seroprevalences were found for RSV, hMPV, and influenza B virus. A high percentage of captive chimpanzees also showed evidence of influenza A H1N1 infections, and had low levels of H3N2 antibodies, while in sera from gorillas and orang-utans antibody levels to influenza A and B viruses were much lower or practically absent. Transmission of respiratory viruses was examined in longitudinal sera of young chimpanzees, and in chimpanzee sera taken during health checks. In young animals isolated cases of influenza infections were monitored, but evidence was found for single introductions followed by a rapid dissemination of RSV and hMPV within the group. Implementation of strict guidelines for handling and housing of nonhuman primates was shown to be an efficient method to reduce the introduction of respiratory infections in colonies of captive animals. RSV seroprevalence rates of chimpanzees remained high, probably due to circulating virus in the chimpanzee colony.
- Published
- 2014
32. Molecular Analysis of a Novel Simian Virus 40 (SV40) Type in Rhesus Macaques and Evidence for Double Infections with the Classical SV40 Type
- Author
-
Sabine van Gessel, Kevin Karremans, Ilona P. Deuzing, Zahra Fagrouch, Henk Niphuis, Willy M. J. M. Bogers, and Ernst J. Verschoor
- Subjects
Microbiology (medical) ,Tumor Virus Infections ,Sequence analysis ,viruses ,Molecular Sequence Data ,Genome, Viral ,Simian virus 40 ,Biology ,Simian ,Virus ,Virology ,Animals ,Enhancer ,Phylogeny ,Polyomavirus Infections ,Incidence ,T-cell receptor ,Primate Diseases ,Sequence Analysis, DNA ,Amplicon ,biology.organism_classification ,Macaca mulatta ,Open reading frame ,Blood ,DNA, Viral ,Leukocytes, Mononuclear ,Lymph Nodes ,Spleen - Abstract
The incidence of simian virus 40 (SV40) infections in rhesus macaques infected with simian-human immunodeficiency viruses (SHIV) and in uninfected animals was determined using PCR. Rates varied from 5% in peripheral blood mononuclear cells of uninfected monkeys to 19.6% in SHIV-infected macaques. Much higher detection rates, up to 75%, were found in lymph nodes and spleen samples of SHIV-infected animals. Sequence analysis of PCR amplicons revealed that they form two genetic clusters, one containing the majority of known SV40 strains and the other formed by variants with 7% genetic difference. Based on this difference, we propose two SV40 types: “type 1” or “classical type” for the majority of SV40 strains and “type 2” for the novel SV40 variants. The genome of one variant, SV40-Ri257, was completely sequenced and analyzed. The agnogene of SV40-Ri257 extends into the VP2 open reading frame and encodes a typical agnoprotein fused to a C-terminal hydrophobic region. The transcriptional control region (TCR) of SV40-Ri257 is the least conserved region compared to type 1 viruses. Particularly, the 3′ end of the TCR, containing the early promoter and enhancer region, exhibits considerable variation. Further analysis of SHIV-infected macaques with type-specific PCRs revealed that the TCR of type 1 was completely conserved, whereas this region in type 2 varied considerably within the early enhancer region. We provide evidence here for the existence of a novel SV40 type in rhesus macaques and show that double infections with both types frequently occur.
- Published
- 2011
- Full Text
- View/download PDF
33. Characterization of novel polyomaviruses from Bornean and Sumatran orang-utans
- Author
-
Jonathan L. Heeney, Sabine van Gessel, Ernst J. Verschoor, Kristin S. Warren, Marlous J. Groenewoud, Henk Niphuis, Zahra Fagrouch, and Aiste Bulavaite
- Subjects
Pongo abelii ,Genes, Viral ,Sequence analysis ,viruses ,Molecular Sequence Data ,Simian ,Polymerase Chain Reaction ,Synteny ,Genome ,Virus ,Serology ,Borneo ,Pongo pygmaeus ,Virology ,biology.animal ,Gene Order ,Animals ,Cluster Analysis ,Primate ,Amino Acid Sequence ,Phylogeny ,Polyomavirus Infections ,biology ,Sequence Analysis, DNA ,biochemical phenomena, metabolism, and nutrition ,Amplicon ,biology.organism_classification ,Ape Diseases ,Tumor Virus Infections ,Indonesia ,DNA, Viral ,African Green Monkey ,Polyomavirus ,Sequence Alignment - Abstract
Serological screening of sera from orang-utans demonstrated a high percentage of sera that cross-reacted with antigens of the polyomavirus (PyV) simian virus 40. Analysis of archival DNA samples from 71 Bornean and eight Sumatran orang-utans with a broad-spectrum PCR assay resulted in the detection of PyV infections in 11 animals from both species. Sequence analysis of the amplicons revealed considerable differences between the PyVs from Bornean and Sumatran orang-utans. The genome from two PyVs, one from each species, was therefore amplified and sequenced. Both viral genomes revealed a characteristic PyV architecture, but lacked an obvious agnogene. Neighbour-joining analysis positioned the viruses in a large cluster together with viruses from bats, bovines, rodents and several primate PyVs from chimpanzees, African green monkeys, squirrel monkeys and the human Merkel cell PyV.
- Published
- 2009
- Full Text
- View/download PDF
34. Molecular characterization of the first polyomavirus from a New World primate: squirrel monkey polyomavirus
- Author
-
Ernst J. Verschoor, Marlous J. Groenewoud, Aruna Kewalapat, Jonathan L. Heeney, Zahra Fagrouch, Sabine van Gessel, and Marja Kik
- Subjects
Squirrel monkey polyomavirus ,viruses ,Molecular Sequence Data ,Genome, Viral ,Genome ,Virus ,Papovaviridae ,chemistry.chemical_compound ,Virology ,biology.animal ,Animals ,Primate ,Amino Acid Sequence ,Saimiri ,Phylogeny ,Binding Sites ,Base Sequence ,biology ,Phylogenetic tree ,Gene Amplification ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,chemistry ,DNA, Viral ,Capsid Proteins ,African Green Monkey ,Polyomavirus ,DNA - Abstract
DNA samples from a variety of New World monkeys were screened by using a broad-spectrum PCR targeting the VP1 gene of polyomaviruses. This resulted in the characterization of the first polyomavirus from a New World primate. This virus naturally infects squirrel monkeys (Saimiri sp.) and is provisionally named squirrel monkey polyomavirus (SquiPyV). The complete genome of SquiPyV is 5075 bp in length, and encodes the small T and large T antigens and the three structural proteins VP1, VP2 and VP3. Interestingly, the late region also encodes a putative agnoprotein, a feature that it shares with other polyomaviruses from humans, baboons and African green monkeys. Comparison with other polyomaviruses revealed limited sequence similarity to any other polyomavirus, and phylogenetic analysis of the VP1 gene confirmed its uniqueness.
- Published
- 2008
- Full Text
- View/download PDF
35. Complete Genome Sequence of a Novel Chimpanzee Polyomavirus from a Western Common Chimpanzee
- Author
-
Ernst J. Verschoor, Zahra Fagrouch, Hester Buitendijk, Ivanela Kondova, Jolanda van Persie, Tom Haaksma, and Willy M. J. M. Bogers
- Subjects
0301 basic medicine ,Whole genome sequencing ,viruses ,virus diseases ,Biology ,biochemical phenomena, metabolism, and nutrition ,Virology ,Virus ,Chimpanzee genome project ,03 medical and health sciences ,030104 developmental biology ,Chimpanzee polyomavirus ,biology.animal ,Viruses ,Genetics ,Common chimpanzee ,Molecular Biology - Abstract
We report here the full-length genome sequence of a novel chimpanzee polyomavirus. Viral sequences were recovered from colon, bladder, and ureter tissue from a western common chimpanzee. The virus is genetically closely related to the human BK polyomavirus.
- Published
- 2016
36. Role of microbial translocation in soluble CD14 up-regulation in HIV-, but not in HCV-, infected chimpanzees
- Author
-
Petra Mooij, Ernst J. Verschoor, Ivonne G. Nieuwenhuis, Andre Boonstra, Gerrit Koopman, Babs E. Verstrepen, Zahra Fagrouch, Ivanela Kondova, and Gastroenterology & Hepatology
- Subjects
0301 basic medicine ,Pan troglodytes ,Lipopolysaccharide ,CD14 ,Hepatitis C virus ,Lipopolysaccharide Receptors ,HIV Infections ,Hepacivirus ,medicine.disease_cause ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Downregulation and upregulation ,SDG 3 - Good Health and Well-being ,Virology ,medicine ,Animals ,Humans ,Intestinal Mucosa ,Pathological ,biology ,virus diseases ,Simian immunodeficiency virus ,Hepatitis C ,Up-Regulation ,Intestines ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Bacterial Translocation ,Immunology ,HIV-1 ,biology.protein ,030211 gastroenterology & hepatology ,Antibody ,Hepatic fibrosis - Abstract
During human immunodeficiency virus (HIV) infection, soluble CD14 (sCD14) is up-regulated as a consequence of pathological disruption of the gut epithelial barrier, and subsequent increased microbial translocation. Also in hepatitis C virus (HCV)-infected patients with advanced liver fibrosis, increased levels of sCD14 have been reported. Since the liver plays an important role in clearance of translocated bacterial products, hepatic fibrosis may negatively affect clearance and thus contribute to higher sCD14 levels. Chimpanzees (Pan troglodytes) infected with HCV typically show no signs of liver fibrosis. Here, we have tested the hypothesis that increased levels of sCD14 occur in the absence of hepatic fibrosis or microbial translocation in chimpanzees chronically infected with HCV. sCD14 was up-regulated in both HIV/simian immunodeficiency virus (SIV)- and HCV-infected chimpanzees. In HIV/SIV-infected chimpanzees, intestinal fatty acid-binding protein, a marker for gut perturbation, lipopolysaccharide (LPS)-binding-protein and LPS core antibodies, confirm that sCD14 up-regulation was caused by increased microbial translocation. In HCV-infected chimpanzees, no evidence was found for increased microbial translocation despite up-regulation of sCD14. Additionally, the impact of liver fibrosis on microbial translocation was addressed by direct comparison of chimpanzees with a high HCV load and human patients with advanced fibrosis. These data suggest that only in a small minority of HCV patients, hepatic fibrosis corroborates microbial translocation.
- Published
- 2016
37. T cell receptor excision circles (TRECs) analysis during acute intrarectal infection of cynomolgus monkeys with pathogenic chimeric simian human immunodeficiency virus
- Author
-
Maria Rosaria Pavone-Cossut, Fausto Titti, Zahra Fagrouch, Roberta Bona, Massimo Ciccozzi, Silvia Baroncelli, Donatella R.M. Negri, Stefania Catone, Viviana Buffa, Jonathan L. Heeney, Pasqualina Leone, Andrea Cara, and Iole Macchia
- Subjects
Cancer Research ,T-Lymphocytes ,T cell ,CD3 ,Molecular Sequence Data ,Simian Acquired Immunodeficiency Syndrome ,Recent Thymic Emigrant ,HIV Infections ,Biology ,Sequence Homology, Nucleic Acid ,Virology ,medicine ,Animals ,Humans ,Gene ,Base Sequence ,Chimera ,T-cell receptor excision circles ,Simian human immunodeficiency virus ,T-cell receptor ,Genes, T-Cell Receptor ,Macaca fascicularis ,Infectious Diseases ,medicine.anatomical_structure ,Immunology ,HIV-1 ,biology.protein ,Simian Immunodeficiency Virus ,DNA, Circular ,CD8 - Abstract
Several studies have shown the importance of evaluating Recent Thymic Emigrants (RTEs) by quantification of T cell receptor-rearrangement excision circles (TRECs), as a measure of de novo T cell generation during human immunodeficiency virus-1 (HIV-1) infection. To determine whether acute viral infection may have an impact on TRECs, cynomolgus monkeys (Macaca fascicularis) were infected intrarectally with simian human immunodeficiency virus (SHIV) 89.6P(cy11) and the number of signal-joint (sj) TRECs was determined in purified CD4+ and CD8+ populations for up to 28 weeks post-infection. Four weeks after infection, TRECs levels significantly decreased in both CD3+ CD4+ and in CD3+ CD8+ T lymphocytes of infected monkeys, whereas they remained unchanged in uninfected animals. This reduction was followed by a progressive TRECs number recovery in CD3+ CD4+ T lymphocytes that positively correlated with changes in the levels of circulating CD3+ CD4+ T cells. In the CD3+ CD8+ T cell subset, TRECs number remained significantly low and inversely correlated with the increase in the percentages of CD3+ CD8+ T cells. These data suggest that SHIV89.6P(cy11) intrarectal infection of cynomolgus monkeys differently affects TRECs content in CD3+ CD4+ and CD3+ CD8+ T cell subsets.
- Published
- 2007
- Full Text
- View/download PDF
38. Characterization ofα-Defensins Plasma Levels inMacaca Fascicularisand Correlations with Virological Parameters during SHIV89.6Pcy11Experimental Infection
- Author
-
Zuleika Michelini, Claudia Rovetto, Pasqualina Leone, Massimo Ciccozzi, Roberto Belli, Jonathan L. Heeney, Donatella R.M. Negri, Zahra Fagrouch, Martina Borghi, Silvia Baroncelli, Andrea Cara, and Stefania Catone
- Subjects
Male ,alpha-Defensins ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Alpha (ethology) ,Viremia ,Virus Replication ,medicine.disease_cause ,Macaque ,Pathogenesis ,Administration, Rectal ,In vivo ,Virology ,biology.animal ,medicine ,Animals ,Infusions, Intravenous ,Defensin ,integumentary system ,biology ,HIV ,respiratory system ,Simian immunodeficiency virus ,medicine.disease ,Disease Models, Animal ,Macaca fascicularis ,Infectious Diseases ,Viral replication ,Female ,Simian Immunodeficiency Virus - Abstract
Alpha-defensins have been shown to inhibit HIV-1 replication in vitro and may contribute to the overall control of viral replication in vivo. In the present work, we quantitatively measured the levels of alpha-defensins in the plasma of healthy and experimentally SHIV-infected Macaca fascicularis (cynomolgus monkeys), an animal model of AIDS pathogenesis and vaccine development. Characterization of physiological plasma alpha-defensins levels was performed in 12 healthy monkeys following longitudinal analysis using an alpha-defensins ELISA kit currently validated for macaque use. Subsequently, alpha-defensins levels were quantitatively measured in 23 cynomolgus monkeys during titration protocols following both the mucosal and systemic routes of infection with the pathogenic SHIV89.6P(cy11). A significant increase in plasma alpha-defensins levels was consistently observed at early time points in all infected animals, regardless of the infection route. Moreover, a positive correlation was observed between viral replication and levels of alpha-defensins during the acute phase of infection. Interestingly, in the animals infected through the mucosal route, alpha-defensins levels remained significantly higher at later time points, up to 19 weeks from the infection, while in cynomolgus infected intravenously, alpha-defensins levels returned to baseline levels by 4 weeks from infection, suggesting that the different route of infection may differently activate the innate immune response.
- Published
- 2007
- Full Text
- View/download PDF
39. Vaccine protection from CD4+ T-cell loss caused by simian immunodeficiency virus (SIV) mac251 is afforded by sequential immunization with three unrelated vaccine vectors encoding multiple SIV antigens
- Author
-
Daniella Mortier, Peter Liljeström, Jonathan L. Heeney, Henk Niphuis, Gerrit Koopman, Sam O. Hofman, Gerd Sutter, Zahra Fagrouch, and S. Norley
- Subjects
viruses ,Simian Acquired Immunodeficiency Syndrome ,Antibodies, Viral ,Semliki Forest virus ,medicine.disease_cause ,Virus ,Interferon-gamma ,chemistry.chemical_compound ,Virology ,medicine ,Animals ,Vaccines, Synthetic ,biology ,SAIDS Vaccines ,Simian immunodeficiency virus ,Vaccine efficacy ,biology.organism_classification ,Macaca mulatta ,CD4 Lymphocyte Count ,Vaccination ,Rhesus macaque ,chemistry ,Immunology ,Lentivirus ,Interleukin-2 ,Immunization ,Interleukin-4 ,Vaccinia - Abstract
Candidate human immunodeficiency virus (HIV) vaccine strategies that induce strong cellular immune responses protect rhesus macaques that are infected with recombinant simian/human immunodeficiency virus SHIV89.6p from acute CD4+ T-cell loss and delay progression to AIDS. However, similar strategies have not proven as efficacious in the simian immunodeficiency virus (SIV)mac model of AIDS, an infection that causes a slow, steady loss of CD4+ T-cell function and numbers in rhesus macaques similar to that caused by HIV-1, the principal cause of AIDS in humans. Efforts to increase vaccine efficacy by repeated boosting with the same vector are quickly limited by rising anti-vector immune responses. Here, the sequential use of three different vectors (DNA, Semliki Forest virus and modified vaccinia virus Ankara) encoding the same SIVmac structural and regulatory antigens was investigated and demonstrated to prevent or slow the loss of CD4+ T-cells after mucosal challenge with the highly pathogenic SIVmac251 strain. Of particular interest was an inverse association between the extent of T-helper 2 cytokine responses and steady-state virus load. Although limited in the number of animals, this study provides important proof of the efficacy of the triple-vector vaccine strategy against chronic, progressive CD4+ T-cell loss in the rigorous SIVmac/rhesus macaque model of AIDS.
- Published
- 2004
- Full Text
- View/download PDF
40. Protective efficacy of a multicomponent vector vaccine in cynomolgus monkeys after intrarectal simian immunodeficiency virus challenge
- Author
-
Federica Crostarosa, Stefano Boros, Stefania Farcomeni, Jonathan L. Heeney, Stephen Norley, Roberto Belli, Donatella R.M. Negri, Massimo Ciccozzi, Zuleika Michelini, Leonardo Sernicola, Maria Grazia Mancini, Fausto Titti, Peter Liljeström, Zahra Fagrouch, Silvia Baroncelli, Maria Teresa Maggiorella, Antonella Comini, and Stefania Catone
- Subjects
Male ,T-Lymphocytes ,viruses ,Genetic Vectors ,Simian Acquired Immunodeficiency Syndrome ,Vaccinia virus ,Lymphocyte Activation ,medicine.disease_cause ,Semliki Forest virus ,Virus ,Viral vector ,chemistry.chemical_compound ,Virology ,medicine ,Animals ,Antigens, Viral ,Immunization Schedule ,AIDS Vaccines ,Recombination, Genetic ,Vaccines, Synthetic ,Attenuated vaccine ,biology ,Vaccination ,Viral Vaccines ,Vector vaccine ,Simian immunodeficiency virus ,biology.organism_classification ,Semliki forest virus ,Disease Models, Animal ,Macaca fascicularis ,chemistry ,Immunology ,Simian Immunodeficiency Virus ,Vaccinia - Abstract
We investigated the protective efficacy of a systemic triple vector (DNA/rSFV/rMVA)-based vaccine against mucosal challenge with pathogenic simian immunodeficiency virus (SIV) in cynomolgus monkeys. Animals were immunized at week 0 with DNA (intradermally), at weeks 8 and 16 with recombinant Semliki Forest virus (rSFV, subcutaneously) and finally, at week 24, with recombinant modified vaccinia virus Ankara strain (rMVA, intramuscularly). Both DNA and recombinant viral vectors expressed a wide range of SIV proteins (Gag, Pol, Tat, Rev, Env and Nef). This immunization strategy elicited cell-mediated rather than humoral responses that were especially increased following the last boost. Upon intrarectal challenge with pathogenic SIVmac251, three of the four vaccinated monkeys dramatically abrogated virus load to undetectable levels up to 41 weeks after challenge. A major contribution to this vaccine effect appeared to be the T-cell-mediated immune response to vaccine antigens (Gag, Rev, Tat, Nef) seen in the early phase of infection in three of the four vaccinated monkeys. Indeed, the frequency of T-cells producing antigen-induced IFN-γmirrored virus clearance in the vaccinated and protected monkeys. These results, reminiscent of the efficacy of live attenuated virus vaccines, suggest that vaccination with a combination of many viral antigens can induce a robust and stable vaccine-induced immunity able to abrogate virus replication.
- Published
- 2004
- Full Text
- View/download PDF
41. Strong vaccine-induced CD8 T-cell responses have cytolytic function in a chimpanzee clearing HCV infection
- Author
-
Natasja G. de Groot, Petra Mooij, Ronald E. Bontrop, Gerrit Koopman, Babs E. Verstrepen, Willy M. J. M. Bogers, Zahra Fagrouch, Jonathan L. Heeney, and Ernst J. Verschoor
- Subjects
Cytotoxicity, Immunologic ,Viral Hepatitis Vaccines ,Pan troglodytes ,medicine.medical_treatment ,Immunology ,Molecular Sequence Data ,Gene Expression ,lcsh:Medicine ,Hepacivirus ,CD8-Positive T-Lymphocytes ,Viral Nonstructural Proteins ,Major histocompatibility complex ,Epitope ,Epitopes ,Immune system ,MHC class I ,medicine ,Cytotoxic T cell ,Animals ,Amino Acid Sequence ,lcsh:Science ,Immune Response ,Immunity to Infections ,Immunity, Cellular ,Multidisciplinary ,biology ,Viral Core Proteins ,Histocompatibility Antigens Class I ,lcsh:R ,Immunity ,Biology and Life Sciences ,Virology ,Hepatitis C ,Acquired Immune System ,Immunity, Humoral ,Cytolysis ,Cytokine ,Immune System ,biology.protein ,Cytokines ,Immunization ,lcsh:Q ,CD8 ,Research Article - Abstract
A single correlate of effective vaccine protection against chronic HCV infection has yet to be defined. In this study, we analyzed T-cell responses in four chimpanzees, immunized with core-E1-E2-NS3 and subsequently infected with HCV1b. Viral clearance was observed in one animal, while the other three became chronically infected. In the animal that cleared infection, NS3-specific CD8 T-cell responses were observed to be more potent in terms of frequency and polyfunctionality of cytokine producing cells. Unique to this animal was the presence of killing-competent CD8 T-cells, specific for NS3 1258-1272, being presented by the chimpanzee MHC class I molecule Patr-A*03∶01, and a high affinity recognition of this epitope. In the animals that became chronically infected, T-cells were able to produce cytokines against the same peptide but no cytolysis could be detected. In conclusion, in the animal that was able to clear HCV infection not only cytokine production was observed but also cytolytic potential against specific MHC class I/peptide-combinations.
- Published
- 2014
42. Vaccine-induced protection of rhesus macaques against plasma viremia after intradermal infection with a European lineage 1 strain of West Nile virus
- Author
-
Herman Oostermeijer, Marina De Filette, Michael S. Diamond, Babs E. Verstrepen, Niek N. Sanders, Linda Stertman, Henk Niphuis, Luisa Barzon, Stefan Chabierski, Willy M. J. M. Bogers, Tom Haaksma, Melanie van Heteren, Sebastian Ulbert, Ernst J. Verschoor, Ivanela Kondova, Zahra Fagrouch, Sofia E. Magnusson, Giorgio Palù, Julianna Lisziewicz, Orsolya Lőrincz, and Publica
- Subjects
CD4-Positive T-Lymphocytes ,viruses ,animal diseases ,lcsh:Medicine ,CD8-Positive T-Lymphocytes ,Antibodies, Viral ,IMMUNOGENICITY ,Viral Envelope Proteins ,ENVELOPE PROTEIN ,Medicine and Health Sciences ,West Nile Virus ,West Nile Virus Vaccines ,lcsh:Science ,PRECLINICAL EVALUATION ,Immunity, Cellular ,Multidisciplinary ,IMMUNE-RESPONSES ,Viral Vaccine ,Immunogenicity ,virus diseases ,Infectious Disease Immunology ,Viral Load ,Vaccination and Immunization ,3. Good health ,Europe ,Vaccination ,REPLIVAX WN ,Viral load ,Research Article ,CYCLE FLAVIVIRUS VACCINE ,Injections, Intradermal ,Immunology ,UNITED-STATES ,Viremia ,CD8(+) T-CELLS ,Biology ,Interferon-gamma ,Vaccine Development ,medicine ,Animals ,Prophylaxis ,lcsh:R ,Biology and Life Sciences ,Outbreak ,Vaccine efficacy ,medicine.disease ,Macaca mulatta ,ENCEPHALITIS-VIRUS ,Virology ,Immunity, Humoral ,Protein Structure, Tertiary ,nervous system diseases ,MICE ,Culicidae ,Clinical Immunology ,Pre-Exposure Prophylaxis ,lcsh:Q ,West Nile Fever - Abstract
The mosquito-borne West Nile virus (WNV) causes human and animal disease with outbreaks in several parts of the world including North America, the Mediterranean countries, Central and East Europe, the Middle East, and Africa. Particularly in elderly people and individuals with an impaired immune system, infection with WNV can progress into a serious neuroinvasive disease. Currently, no treatment or vaccine is available to protect humans against infection or disease. The goal of this study was to develop a WNV-vaccine that is safe to use in these high-risk human target populations. We performed a vaccine efficacy study in non-human primates using the contemporary, pathogenic European WNV genotype 1a challenge strain, WNV-Ita09. Two vaccine strategies were evaluated in rhesus macaques (Macaca mulatta) using recombinant soluble WNV envelope (E) ectodomain adjuvanted with Matrix-M, either with or without DNA priming. The DNA priming immunization was performed with WNV-DermaVir nanoparticles. Both vaccination strategies successfully induced humoral and cellular immune responses that completely protected the macaques against the development of viremia. In addition, the vaccine was well tolerated by all animals. Overall, The WNV E protein adjuvanted with Matrix-M is a promising vaccine candidate for a non-infectious WNV vaccine for use in humans, including at-risk populations.
- Published
- 2014
43. DNA/long peptide vaccination against conserved regions of SIV induces partial protection against SIVmac251 challenge
- Author
-
Petra Mooij, Ronald E. Bontrop, Niels Beenhakker, Jan W. Drijfhout, Josef Koestler, Zahra Fagrouch, Ralf Wagner, Tomáš Hanke, Willy M. J. M. Bogers, Gaby G. M. Doxiadis, Cornelis J. M. Melief, Gerrit Koopman, Ivonne G. Nieuwenhuis, and Ernst J. Verschoor
- Subjects
SIVmac251 ,Immunogen ,HIV vaccine ,macaques ,Immunology ,Simian Acquired Immunodeficiency Syndrome ,Biology ,Viral vector ,Adjuvants, Immunologic ,Interferon ,MHC class I ,medicine ,Vaccines, DNA ,Immunology and Allergy ,Animals ,Vaccines, Synthetic ,Vaccination ,SAIDS Vaccines ,Virology ,Macaca mulatta ,synthetic long peptides ,Infectious Diseases ,Vaccines, Subunit ,Peptide vaccine ,biology.protein ,conserved regions ,CD8 ,medicine.drug - Abstract
OBJECTIVES: We recently developed a HIVconsv vaccine strategy, consisting of combined conserved regions of HIV-1, to adequately cover viral diversity. To evaluate efficacy in nonhuman primates, an equivalent SIV-derived immunogen SIVconsv was designed and delivered as plasmid DNA or synthetic long peptides. DESIGN: Rhesus macaques lacking protective MHC class I alleles Mamu-A*001 : 01, B*008 : 01, B*017 : 01 were immunized with either SIVconsv synthetic long peptides (S) alone or in combination with plasmid DNA encoding the same conserved regions (D) using SSS or DDSS regimens. METHODS: The SIVconsv synthetic long peptide vaccine consisted of 46 approximately 30-amino acid-long peptides emulsified in Montanide ISA-720 and adjuvanted with pegylated type I interferon and imiquimod. RESULTS: Both SSS and DDSS regimens generated high frequencies of SIV-specific IFN-γ-producing cells comparable with reported adenoviral vector systems. Strong polyfunctional CD4⁺ T-cell and modest CD8⁺ T-cell responses were generated, which were of central memory T-cell phenotype. Furthermore, SIVconsv-specific antibody responses were induced capable of recognizing the Env glycoprotein. Eight weeks after the last immunization, control and SIVconsv-vaccinated animals were challenged intrarectally with 10 MID50 of pathogenic SIVmac251. Two out of six animals in the DDSS group were protected against infection, while all 14 animals in the SSS and two control groups were infected. Vaccine induced SIV-specific IgG responses in mucosal washes prechallenge were highest in the two protected animals. CONCLUSION: This study demonstrates that vaccine-elicited responses towards conserved regions can afford partial protection against a high-dose intrarectal SIVmac251 challenge.
- Published
- 2013
44. Novel polyomaviruses in South American bats and their relationship to other members of the family Polyomaviridae
- Author
-
Roya Sarwari, Anne Lavergne, Benoit de Thoisy, Vincent Lacoste, Marguerite Delaval, Ernst J. Verschoor, Zahra Fagrouch, Biomedical Primate Research Centre [Rijswijk] (BPRC), Laboratoire des Interactions Virus-Hôtes [Cayenne, Guyane Française], Institut Pasteur de la Guyane, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP), Service développement Sylvétude [Réserve Montabo, Cayenne], ONF - Direction régionale de la Guyane [Cayenne], Office National des Forêts (ONF)-Office National des Forêts (ONF), and European Community Research Infrastructures Program grant RII3-CT-2006-026155 and Fonds Européen de développement régional
- Subjects
viruses ,MESH: Polyomaviridae/genetics ,Merkel cell polyomavirus ,MESH: Pongo/virology ,Genome ,Genus ,Chiroptera ,MESH: Animals ,MESH: Phylogeny ,Phylogeny ,0303 health sciences ,MESH: Disease Reservoirs/virology ,MESH: Gorilla gorilla/virology ,Phylogenetic tree ,MESH: Polyomavirus/classification ,[SDV.BID.EVO]Life Sciences [q-bio]/Biodiversity/Populations and Evolution [q-bio.PE] ,virus diseases ,3. Good health ,French Guiana ,MESH: Polyomaviridae/isolation & purification ,South american ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,MESH: Polyomaviridae/classification ,MESH: Genome, Viral ,Polyomaviridae ,Polyomavirus ,Family Polyomaviridae ,animal structures ,Pan troglodytes ,Molecular Sequence Data ,Zoology ,Genome, Viral ,Biology ,MESH: Pan troglodytes/virology ,Virus ,03 medical and health sciences ,Species Specificity ,Virology ,MESH: French Guiana ,MESH: Polyomavirus/isolation & purification ,MESH: Polyomavirus/genetics ,MESH: Species Specificity ,Animals ,Humans ,Natural reservoir ,MESH: Chiroptera/virology ,MESH: Disease Reservoirs/veterinary ,030304 developmental biology ,Disease Reservoirs ,MESH: Molecular Sequence Data ,MESH: Humans ,Gorilla gorilla ,030306 microbiology ,Pongo ,MESH: South America ,biochemical phenomena, metabolism, and nutrition ,South America ,biology.organism_classification ,MESH: Merkel cell polyomavirus/classification ,MESH: Merkel cell polyomavirus/genetics - Abstract
International audience; Bats are the natural reservoir of a variety of viruses, including a polyomavirus from a North American brown bat. We investigated 163 spleen samples from 22 bat species from French Guiana for the presence of polyomaviruses. In total, we detected 25 positive animals belonging to nine different bat species. Phylogenetic analysis was performed on the genomes of eight representative polyomaviruses, and showed that the bat polyomaviruses form three distinct lineages within the Orthopolyomavirus genus, and are genetically different from the previously described North American bat virus. Interestingly, two lineages cluster with polyomaviruses found in chimpanzees, orangutans and gorillas. In addition, one group of bat polyomaviruses is genetically related to the human Merkel cell polyomavirus.
- Published
- 2012
- Full Text
- View/download PDF
45. Protection in macaques immunized with HIV-1 candidate vaccines can be predicted using the kinetics of their neutralizing antibodies
- Author
-
Wim Koornstra, Zahra Fagrouch, Jonathan L. Heeney, David Davis, Ernst J. Verschoor, Daniella Mortier, and Willy M. J. M. Bogers
- Subjects
lcsh:Medicine ,Neutralization ,0302 clinical medicine ,Immunodeficiency Viruses ,030212 general & internal medicine ,Neutralizing antibody ,lcsh:Science ,AIDS Vaccines ,0303 health sciences ,Multidisciplinary ,Viral Vaccine ,Vaccination ,Animal Models ,3. Good health ,AIDS ,Medicine ,Infectious diseases ,Viral load ,Macaque ,Research Article ,Immunology ,HIV prevention ,Sexually Transmitted Diseases ,Immunoglobulins ,Viremia ,Viral diseases ,Biology ,Microbiology ,Virus ,Immunomodulation ,03 medical and health sciences ,Model Organisms ,Antigen ,Virology ,Vaccine Development ,medicine ,Animals ,030304 developmental biology ,lcsh:R ,Immunity ,HIV ,Viral Vaccines ,medicine.disease ,Antibodies, Neutralizing ,Macaca mulatta ,Kinetics ,biology.protein ,HIV-1 ,Clinical Immunology ,lcsh:Q - Abstract
Background A vaccine is needed to control the spread of human immunodeficiency virus type 1 (HIV-1). An in vitro assay that can predict the protection induced by a vaccine would facilitate the development of such a vaccine. A potential candidate would be an assay to quantify neutralization of HIV-1. Methods and Findings We have used sera from rhesus macaques that have been immunized with HIV candidate vaccines and subsequently challenged with simian human immunodeficiency virus (SHIV). We compared neutralization assays with different formats. In experiments with the standardized and validated TZMbl assay, neutralizing antibody titers against homologous SHIVSF162P4 pseudovirus gave a variable correlation with reductions in plasma viremia levels. The target cells used in the assays are not just passive indicators of virus infection but are actively involved in the neutralization process. When replicating virus was used with GHOST cell assays, events during the absorption phase, as well as the incubation phase, determine the level of neutralization. Sera that are associated with protection have properties that are closest to the traditional concept of neutralization: the concentration of antibody present during the absorption phase has no effect on the inactivation rate. In GHOST assays, events during the absorption phase may inactivate a fixed number, rather than a proportion, of virus so that while complete neutralization can be obtained, it can only be found at low doses particularly with isolates that are relatively resistant to neutralization. Conclusions Two scenarios have the potential to predict protection by neutralizing antibodies at concentrations that can be induced by vaccination: antibodies that have properties close to the traditional concept of neutralization may protect against a range of challenge doses of neutralization sensitive HIV isolates; a window of opportunity also exists for protection against isolates that are more resistant to neutralization but only at low challenge doses.
- Published
- 2011
46. Seroprevalence of SV40-like polyomavirus infections in captive and free-ranging macaque species
- Author
-
Ernst J. Verschoor, Jonathan L. Heeney, M.C. Pizarro, Henk Niphuis, P. Christian, Zahra Fagrouch, and K. Sasnauskas
- Subjects
Polyomavirus Infections ,General Veterinary ,Free ranging ,biology ,animal diseases ,Macaca sylvanus ,Animals, Wild ,Simian ,biology.organism_classification ,Antibodies, Viral ,Macaque ,Virology ,Virus ,Seroepidemiologic Studies ,biology.animal ,biology.protein ,Seroprevalence ,Animals ,Macaca ,Mauritius ,Animal Science and Zoology ,Animals, Zoo ,Capsid Proteins ,Antibody - Abstract
Background and methods To investigate the seroprevalence of polyomavirus infections in macaques, we analyzed 1579 sera from nine different species for antibodies cross-reactive with simian virus 40 (SV40) in an enzyme-linked immunosorbent assay. Most samples were collected from captive animals, but we also investigated a colony of free-ranging Barbary macaques (Macaca sylvanus). Results High seropositive rates were found in rhesus macaques (Macaca mulatta; 74.7%), cynomolgus macaques (Macaca fascicularis; 44.8%) and Tonkean macaques (Macaca tonkeana; 41.7%), especially in animals imported from China. Low rates were measured in cynomolgus macaques from Mauritius (8.8%), and in Barbary macaques (1.4%). Seropositivity was age-dependent increasing to >70% in animals of 5 years and older. Conclusions High seroprevalence rates were found in different species of macaques, dependent on their origin. Very low infection rates found in Barbary macaques and cynomolgus macaques from Mauritius suggest that these animals in the wild are not commonly infected by SV40-like viruses.
- Published
- 2008
47. Vaccine-induced early control of hepatitis C virus infection in chimpanzees fails to impact on hepatic PD-1 and chronicity
- Author
-
Christine S, Rollier, Glaucia, Paranhos-Baccala, Ernst J, Verschoor, Babs E, Verstrepen, Joost A R, Drexhage, Zahra, Fagrouch, Jean-Luc, Berland, Florence, Komurian-Pradel, Blandine, Duverger, Nourredine, Himoudi, Caroline, Staib, Marcus, Meyr, Mike, Whelan, Joseph A, Whelan, Victoria C, Adams, Victoria A, Adams, Esther, Larrea, José I, Riezu, Juan J, Lasarte, Juan José, Lasarte, Birke, Bartosch, Francois-L, Cosset, Willy J M, Spaan, Helmut M, Diepolder, Gerd R, Pape, Gerd, Sutter, Genevieve, Inchauspe, and Jonathan L, Heeney
- Subjects
Viral Hepatitis Vaccines ,Pan troglodytes ,Hepatitis C virus ,Programmed Cell Death 1 Receptor ,Hepacivirus ,Biology ,CD8-Positive T-Lymphocytes ,medicine.disease_cause ,Virus ,Immune system ,Antigen ,Antigens, CD ,medicine ,Animals ,Antigens, Viral ,B-Lymphocytes ,Hepatology ,Viral Load ,Virology ,Hepatitis C ,Vaccination ,Chronic infection ,Immunization ,Immunology ,Chronic Disease ,DNA, Viral ,Cytokines ,Viral disease ,Apoptosis Regulatory Proteins - Abstract
UNLABELLED: Broad T cell and B cell responses to multiple HCV antigens are observed early in individuals who control or clear HCV infection. The prevailing hypothesis has been that similar immune responses induced by prophylactic immunization would reduce acute virus replication and protect exposed individuals from chronic infection. Here, we demonstrate that immunization of naïve chimpanzees with a multicomponent HCV vaccine induced robust HCV-specific immune responses, and that all vaccinees exposed to heterologous chimpanzee-adapted HCV 1b J4 significantly reduced viral RNA in serum by 84%, and in liver by 99% as compared to controls (P=0.024 and 0.028, respectively). However, despite control of HCV in plasma and liver in the acute period, in the chronic phase, 3 of 4 vaccinated animals developed persistent infection. Analysis of expression levels of proinflammatory cytokines in serial hepatic biopsies failed to reveal an association with vaccine outcome. However, expression of IDO, CTLA-4 [corrected] and PD-1 levels in liver correlated with clearance or chronicity. CONCLUSION: Despite early control of virus load, a virus-associated tolerogenic-like state can develop in certain individuals independent of vaccination history.
- Published
- 2007
48. Sustained conservation of CD4+ T cells in multiprotein triple modality-immunized rhesus macaques after intrarectal challenge with simian immunodeficiency virus
- Author
-
Ulrike Sauermann, Gerhard Hunsmann, Jonathan L. Heeney, Nicole Stolte-Leeb, Christiane Stahl-Hennig, Stephen Norley, Monika Franz, and Zahra Fagrouch
- Subjects
CD4-Positive T-Lymphocytes ,Modified vaccinia Ankara ,viruses ,Immunology ,Genetic Vectors ,Immunization, Secondary ,Simian Acquired Immunodeficiency Syndrome ,Vaccinia virus ,Semliki Forest virus ,medicine.disease_cause ,Antibodies, Viral ,Antigen ,Administration, Rectal ,Virology ,medicine ,Vaccines, DNA ,Animals ,biology ,SAIDS Vaccines ,Simian immunodeficiency virus ,biology.organism_classification ,Macaca mulatta ,Semliki forest virus ,Regimen ,Immunization ,biology.protein ,Molecular Medicine ,Simian Immunodeficiency Virus ,Antibody ,Viral load - Abstract
As part of a European multicenter study designed to determine the optimal combination and order of a mixed-modality vaccine against acquired immunodeficiency syndrome, rhesus monkeys received a combination of three different vectors, all expressing the same Simian Immunodeficiency Virus (SIV) genes followed by mucosal challenge with highly pathogenic SIV. In the study reported here, animals were primed with DNA followed by one booster immunization with Semliki Forest Virus (SFV) and two immunizations with modified Vaccinia Ankara (MVA). To address the relevance of mucosal immunization, we compared systemic versus a combination of systemic and mucosal antigen application. Although all vaccinees became infected after intrarectal challenge with SIV, most (six of eight) were protected from profound loss of CD4+ cells. In addition, vaccinees showed lower viral loads than did controls (p < 0.05). Overall, these protective effects were more pronounced in those animals whose schedule included immunization via the mucosa. In summary, the vaccine regimen used here achieved one important criterion of efficacy: the suppression of disease development as indicated by conservation of CD4+ cells.
- Published
- 2006
49. Phylogeny of primate T lymphotropic virus type 1 (PTLV-1) including various new Asian and African non-human primate strains
- Author
-
Anne-Mieke Vandamme, Ernst J. Verschoor, Zahra Fagrouch, and Sonia Van Dooren
- Subjects
Microbiology (medical) ,Primates ,Cercopithecus ascanius ,Asia ,Primate T-lymphotropic virus 1 ,Molecular Sequence Data ,Zoology ,Gorilla ,Subspecies ,Microbiology ,Macaque ,Genes, env ,Monophyly ,Western lowland gorilla ,biology.animal ,Zoonoses ,Genetics ,Animals ,Primate ,Clade ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Phylogeny ,Human T-lymphotropic virus 1 ,Deltaretrovirus Infections ,biology ,Primate Diseases ,Terminal Repeat Sequences ,biology.organism_classification ,Infectious Diseases ,Africa ,Simian T-lymphotropic virus 1 - Abstract
To further unravel intra- and interspecies PTLV-1 evolution in Asia and Africa, we phylogenetically analysed 15 new STLV-1 LTR and env sequences discovered in eight different Asian and African non-human primate species. We show that orang-utan STLV-1s form a tight, deeply branching monophyletic cluster between Asian STLV-1 macaque species clades, suggesting natural cross-species transmission. Novel viruses of Macaca maura, Macaca nigra and siamang cluster with other Sulawesian STLV-1s, demonstrating close relatedness among the STLV-1s in these insular species and suggesting cross-species transmission to a siamang in captivity. Viruses from Western chimpanzees and a Western lowland gorilla cluster within the HTLV-lb/STLV-1 clade, the latter close to a human strain, indicative of zoonosis. A new STLV-1 from Cercopithecus ascanius differs from the published STLV-Cas57, explainable by the existence of five geographically separated subspecies. Barbary macaques, not yet described to be STLV-infected, carry a relatively recent acquired, typical African STLV-1, giving us no clue on the phylogeographical origin of PTLV-1.
- Published
- 2006
50. Reduction of viral loads by multigenic DNA priming and adenovirus boosting in the SIVmac-macaque model
- Author
-
You S. Suh, Stephen Norley, Zahra Fagrouch, Monika Franz, Gerhard Hunsmann, Ki Seok Park, Young Chul Sung, Jonathan L. Heeney, Ulrike Sauermann, Christiane Stahl-Hennig, Doris Wilfingseder, and Heribert Stoiber
- Subjects
viruses ,animal diseases ,T-Lymphocytes ,Genetic Vectors ,Immunization, Secondary ,Simian Acquired Immunodeficiency Syndrome ,Viral Nonstructural Proteins ,medicine.disease_cause ,Antibodies, Viral ,Macaque ,Injections, Intramuscular ,DNA vaccination ,Viral vector ,Adenoviridae ,Interferon-gamma ,Adjuvants, Immunologic ,Neutralization Tests ,biology.animal ,medicine ,Vaccines, DNA ,Animals ,Viremia ,Neutralizing antibody ,Viral Structural Proteins ,General Veterinary ,General Immunology and Microbiology ,biology ,ELISPOT ,Vaccination ,Public Health, Environmental and Occupational Health ,SAIDS Vaccines ,virus diseases ,Simian immunodeficiency virus ,Viral Load ,biology.organism_classification ,Virology ,Interleukin-12 ,Macaca mulatta ,CD4 Lymphocyte Count ,Infectious Diseases ,Lentivirus ,Immunology ,biology.protein ,Molecular Medicine ,Simian Immunodeficiency Virus ,Plasmids - Abstract
In this study, we investigated the ability of a multigenic SIV DNA prime/replication-defective adenovirus serotype 5 (rAd/SIV) boost regimen to induce SIV-specific immune responses and protection against intrarectal challenge with SIVmac251 in rhesus macaques. Four rhesus macaques were immunized intramuscularly three times at 8-week intervals with SIV DNA vaccine and boosted once with rAd/SIV vaccine Four control macaques received the same amount of mock plasmid DNA and mock adenovirus vector. While the SIV DNA vaccine included plasmids expressing a mutated human IL-12 gene (IL-12N222L) as well as SIVmac239 structural and regulatory genes, the rAd/SIV vaccine contained rAd vectors expressing SIVmac239 genes only. Immunization with SIV DNA vaccine alone induced SIV-specific IFN-γ ELISPOT responses in only two of four vaccinated macaques, whereas all animals developed SIV-specific T-cell responses and Env- and Tat-specific antibody responses following the rAd/SIV vaccine boost. Upon intrarectal challenge with pathogenic SIVmac251, strong anamnestic Env-specific binding and neutralizing antibody responses were detected in the vaccinated macaques. Overall, the immunized macaques had lower peak and set-point viral loads than control macaques, suggesting that the induced immune responses play a role in the control of viremia. In addition, the loss of CD4 + T cells was delayed in the vaccinated macaques after challenge. These results indicate that the multigenic DNA prime-adenovirus boost immunization may be a promising approach in developing an effective AIDS vaccine.
- Published
- 2005
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.