Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 19(9): 1035, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29955109

ABSTRACT

In the version of this article initially published, the accession code for the RNA-seq data set deposited in the NCBI public repository Sequence Read Archive was missing from the 'Data availability' subsection of the Methods section. The accession code is SRP125477.

2.
Nat Immunol ; 18(12): 1310-1320, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29035391

ABSTRACT

The hygiene hypothesis postulates that the recent increase in allergic diseases such as asthma and hay fever observed in Western countries is linked to reduced exposure to childhood infections. Here we investigated how infection with a gammaherpesvirus affected the subsequent development of allergic asthma. We found that murid herpesvirus 4 (MuHV-4) inhibited the development of house dust mite (HDM)-induced experimental asthma by modulating lung innate immune cells. Specifically, infection with MuHV-4 caused the replacement of resident alveolar macrophages (AMs) by monocytes with regulatory functions. Monocyte-derived AMs blocked the ability of dendritic cells to trigger a HDM-specific response by the TH2 subset of helper T cells. Our results indicate that replacement of embryonic AMs by regulatory monocytes is a major mechanism underlying the long-term training of lung immunity after infection.


Subject(s)
Asthma/therapy , Macrophages, Alveolar/immunology , Monocytes/immunology , Pyroglyphidae/immunology , Rhadinovirus/immunology , Th2 Cells/immunology , Adoptive Transfer , Animals , Asthma/immunology , Cell Line , Cricetinae , Dendritic Cells/immunology , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Macrophages, Alveolar/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Th2 Cells/transplantation
3.
Proc Natl Acad Sci U S A ; 121(13): e2306763121, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38498711

ABSTRACT

Lactate-proton symporter monocarboxylate transporter 1 (MCT1) facilitates lactic acid export from T cells. Here, we report that MCT1 is mandatory for the development of virus-specific CD8+ T cell memory. MCT1-deficient T cells were exposed to acute pneumovirus (pneumonia virus of mice, PVM) or persistent γ-herpesvirus (Murid herpesvirus 4, MuHV-4) infection. MCT1 was required for the expansion of virus-specific CD8+ T cells and the control of virus replication in the acute phase of infection. This situation prevented the subsequent development of virus-specific T cell memory, a necessary step in containing virus reactivation during γ-herpesvirus latency. Instead, persistent active infection drove virus-specific CD8+ T cells toward functional exhaustion, a phenotype typically seen in chronic viral infections. Mechanistically, MCT1 deficiency sequentially impaired lactic acid efflux from activated CD8+ T cells, caused an intracellular acidification inhibiting glycolysis, disrupted nucleotide synthesis in the upstream pentose phosphate pathway, and halted cell proliferation which, ultimately, promoted functional CD8+ T cell exhaustion instead of memory development. Taken together, our data demonstrate that MCT1 expression is mandatory for inducing T cell memory and controlling viral infection by CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Symporters , Animals , Mice , CD8-Positive T-Lymphocytes/metabolism , Lactic Acid/metabolism , Biological Transport , Symporters/genetics , Symporters/metabolism
4.
Immunity ; 46(3): 457-473, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28329706

ABSTRACT

Living in a microbe-rich environment reduces the risk of developing asthma. Exposure of humans or mice to unmethylated CpG DNA (CpG) from bacteria reproduces these protective effects, suggesting a major contribution of CpG to microbe-induced asthma resistance. However, how CpG confers protection remains elusive. We found that exposure to CpG expanded regulatory lung interstitial macrophages (IMs) from monocytes infiltrating the lung or mobilized from the spleen. Trafficking of IM precursors to the lung was independent of CCR2, a chemokine receptor required for monocyte mobilization from the bone marrow. Using a mouse model of allergic airway inflammation, we found that adoptive transfer of IMs isolated from CpG-treated mice recapitulated the protective effects of CpG when administered before allergen sensitization or challenge. IM-mediated protection was dependent on IL-10, given that Il10-/- CpG-induced IMs lacked regulatory effects. Thus, the expansion of regulatory lung IMs upon exposure to CpG might underlie the reduced risk of asthma development associated with a microbe-rich environment.


Subject(s)
Chemotaxis, Leukocyte/immunology , DNA, Bacterial/immunology , Hypersensitivity/immunology , Macrophages, Alveolar/immunology , Respiratory Hypersensitivity/immunology , Animals , Disease Models, Animal , Flow Cytometry , Macrophage Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodeoxyribonucleotides/immunology , Spleen/immunology
5.
J Virol ; 97(7): e0013523, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37338377

ABSTRACT

The development of effective and flexible vaccine platforms is a major public health challenge, especially in the context of influenza vaccines that have to be renewed every year. Adenoviruses (AdVs) are easy to produce and have a good safety and efficacy profile when administered orally, as demonstrated by the long-term use of oral AdV-4 and -7 vaccines in the U.S. military. These viruses therefore appear to be the ideal backbone for the development of oral replicating vector vaccines. However, research into these vaccines is limited by the ineffectiveness of human AdV replication in laboratory animals. The use of mouse AdV type 1 (MAV-1) in its natural host allows infection to be studied under replicating conditions. Here, we orally vaccinated mice with a MAV-1 vector expressing influenza hemagglutinin (HA) to assess the protection conferred against an intranasal challenge of influenza. We showed that a single oral immunization with this vaccine generates influenza-specific and -neutralizing antibodies and completely protects mice against clinical signs and viral replication, similar to traditional inactivated vaccines. IMPORTANCE Given the constant threat of pandemics and the need for annual vaccination against influenza and possibly emerging agents such as SARS-CoV-2, new types of vaccines that are easier to administer and therefore more widely accepted are a critical public health need. Here, using a relevant animal model, we have shown that replicative oral AdV vaccine vectors can help make vaccination against major respiratory diseases more available, better accepted, and therefore more effective. These results could be of major importance in the coming years in the fight against seasonal or emerging respiratory diseases such as COVID-19.


Subject(s)
Adenoviridae Infections , Adenovirus Vaccines , COVID-19 , Influenza Vaccines , Influenza, Human , Humans , Mice , Animals , Adenoviridae/genetics , Influenza, Human/prevention & control , Antibodies, Viral , SARS-CoV-2 , Immunization , Vaccination/methods , Hemagglutinin Glycoproteins, Influenza Virus/genetics
6.
Virol J ; 21(1): 40, 2024 02 10.
Article in English | MEDLINE | ID: mdl-38341597

ABSTRACT

Since the onset of the coronavirus disease (COVID-19) pandemic in Belgium, UZ/KU Leuven has played a crucial role as the National Reference Centre (NRC) for respiratory pathogens, to be the first Belgian laboratory to develop and implement laboratory developed diagnostic assays for SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) and later to assess the quality of commercial kits. To meet the growing demand for decentralised testing, both clinical laboratories and government-supported high-throughput platforms were gradually deployed across Belgium. Consequently, the role of the NRC transitioned from a specialised testing laboratory to strengthening capacity and coordinating quality assurance. Here, we outline the measures taken by the NRC, the national public health institute Sciensano and the executing clinical laboratories to ensure effective quality management of molecular testing throughout the initial two years of the pandemic (March 2020 to March 2022).


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , COVID-19/diagnosis , COVID-19/epidemiology , Belgium/epidemiology , COVID-19 Testing , Pandemics , Clinical Laboratory Techniques , Molecular Diagnostic Techniques
7.
Nano Lett ; 21(1): 847-853, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33373240

ABSTRACT

Viruses are one of the most efficient pathogenic entities on earth, resulting from millions of years of evolution. Each virus particle carries the minimum number of genes and proteins to ensure their reproduction within host cells, hijacking some host replication machinery. However, the role of some viral proteins is not yet unraveled, with some appearing even redundant. For example, murid herpesvirus 4, the current model for human gammaherpesvirus infection, can bind to cell surface glycosaminoglycans using both glycoproteins gp70 and gH/gL. Here, using atomic force microscopy, we discriminate their relative contribution during virus binding to cell surface glycosaminoglycans. Single-virus force spectroscopy experiments demonstrate that gH/gL is the main actor in glycosaminoglycan binding, engaging more numerous and more stable interactions. We also demonstrated that Fab antibody fragments targeting gH/gL or gp70 appear to be a promising treatment to prevent the attachment of virions to cell surfaces.


Subject(s)
Viral Envelope Proteins , Viruses , Cell Line , Glycoproteins , Humans , Spectrum Analysis
8.
Eur J Immunol ; 49(7): 1067-1081, 2019 07.
Article in English | MEDLINE | ID: mdl-30919955

ABSTRACT

Alternatively activated Mφs (AAMφ) accumulate in hepatic granulomas during schistosomiasis and have been suggested to originate in the bone marrow. What is less understood is how these Mφ responses are regulated after S. mansoni infection. Here, we investigated the role of IL-4 receptor α-chain (IL-4Rα)-signalling in the dynamics of liver Mφ responses. We observed that IL-4Rα signalling was dispensable for the recruitment of Ly6Chi monocytes and for their conversion into F4/80hi CD64hi CD11bhi Mφ. Moreover, while IL-4Rα provided an AAMφ phenotype to liver F4/80hi CD64hi CD11bhi Mφ that was associated with regulation of granuloma formation, it was dispensable for host survival. Resident F4/80hi CD64hi CD11blo Mφ did not upregulate the AAMφ signature gene Ym1. Rather, resident Mφ nearly disappeared by week 8 after infection and artificial ablation of resident Mφ in CD169DTR mice did not affect the response to S. mansoni infection. Interestingly, ablation of CD169+ cells in naive mice resulted in the accumulation of F4/80hi CD64hi CD11bhi Mφ, which was amplified when ablation occurred during schistosomiasis. Altogether, our results suggest the ablation of resident KCs after S. mansoni infection to be associated with the recruitment and accumulation of F4/80hi CD64hi CD11bhi Mφ with lyz2-dependent IL-4Rα contributing to the regulation of granuloma inflammation but being dispensable for host survival.


Subject(s)
Granuloma/immunology , Kupffer Cells/immunology , Liver/pathology , Macrophages/immunology , Receptors, Cell Surface/metabolism , Schistosoma mansoni/physiology , Schistosomiasis/immunology , Ablation Techniques , Animals , Disease Models, Animal , Female , Humans , Macrophage Activation , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, Cell Surface/genetics , Signal Transduction
9.
J Virol ; 93(13)2019 07 01.
Article in English | MEDLINE | ID: mdl-30996103

ABSTRACT

Since the 1970s, replication-competent human adenoviruses 4 and 7 have been used as oral vaccines to protect U.S. soldiers against the severe respiratory diseases caused by these viruses. These vaccines are thought to establish a digestive tract infection conferring protection against respiratory challenge through antibodies. The success of these vaccines makes replication-competent adenoviruses attractive candidates for use as oral vaccine vectors. However, the inability of human adenoviruses to replicate efficiently in laboratory animals has hampered the study of such vectors. Here, we used mouse adenovirus type 1 (MAV-1) in mice to study oral replication-competent adenovirus-based vaccines. We show that MAV-1 oral administration provides protection that recapitulates the protection against homologous respiratory challenge observed with adenovirus 4 and 7 vaccines. Moreover, live oral MAV-1 vaccine better protected against a respiratory challenge than inactivated vaccines. This protection was linked not only with the presence of MAV-1-specific antibodies but also with a better recruitment of effector CD8 T cells. However, unexpectedly, we found that such oral replication-competent vaccine systemically spread all over the body. Our results therefore support the use of MAV-1 to study replication-competent oral adenovirus-based vaccines but also highlight the fact that those vaccines can disseminate widely in the body.IMPORTANCE Replication-competent adenoviruses appear to be promising vectors for the development of oral vaccines in humans. However, the study and development of these vaccines suffer from the lack of any reliable animal model. In this study, mouse adenovirus type 1 was used to develop a small-animal model for oral replication-competent adenovirus vaccines. While this model reproduced in mice what is observed with human adenovirus oral vaccines, it also highlighted that oral immunization with such a replication-competent vaccine is associated with the systemic spread of the virus. This study is therefore of major importance for the future development of such vaccine platforms and their use in large human populations.


Subject(s)
Adenoviridae Infections/prevention & control , Adenovirus Vaccines/immunology , Administration, Oral , Gastrointestinal Tract/immunology , Vaccination , Adenoviridae/immunology , Adenoviridae Infections/immunology , Adenoviruses, Human , Animals , Antibodies, Viral/immunology , Disease Models, Animal , Female , Humans , Immunization , Lung/pathology , Mice , Mice, Inbred BALB C , Vaccines, Attenuated/immunology , Vaccines, Synthetic/immunology
10.
J Antimicrob Chemother ; 73(8): 2095-2103, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29788354

ABSTRACT

Objectives: To investigate the efficacy of cidofovir to block gammaherpesvirus replication in the context of sexual transmission. Methods: A luciferase-expressing strain of murid herpesvirus 4 (MuHV-4) was used to monitor genital virus excretion from infected female BALB/c mice and sexual transmission to naive males. The efficiency of cidofovir to block genital excretion from infected females or replication and host colonization of naive males after sexual contact was tested by treating infected females (either once daily or at a single timepoint), naive males before exposure (either once daily or at a single timepoint) or males 24 h post-exposure. Results: We showed that daily treatment of infected females can reduce MuHV-4 genital shedding by 75%. Similarly, daily preventive treatment of naive males was sufficient to block viral replication and latency establishment in males. In contrast, a single administration of cidofovir to infected females at day 14 post-infection or to naive males 2 to 6 days before contact with MuHV-4-excreting females was not sufficient to significantly reduce viral shedding from females or infection of males, respectively. Interestingly, a single administration of cidofovir to males 24 h after contact with MuHV-4-infected females excreting the virus in the genital tract significantly reduced virus replication in males and seroconversion. Conclusions: Altogether, our results show that cidofovir can significantly reduce gammaherpesvirus replication, excretion and colonization of the naive partner in the context of sexual transmission. Such treatments could therefore be recommended in some specific conditions where gammaherpesvirus infections could be deleterious.


Subject(s)
Antiviral Agents/pharmacology , Cidofovir/pharmacology , Herpesviridae Infections/drug therapy , Herpesviridae Infections/transmission , Rhadinovirus/drug effects , Sexually Transmitted Diseases, Viral/drug therapy , Animals , Female , Male , Mice, Inbred BALB C , Nucleosides/analogs & derivatives , Post-Exposure Prophylaxis , Rhadinovirus/physiology , Virus Latency , Virus Replication , Virus Shedding
11.
J Virol ; 91(13)2017 07 01.
Article in English | MEDLINE | ID: mdl-28424280

ABSTRACT

Gammaherpesviruses are important human and animal pathogens. Infection control has proven difficult because the key process of transmission is ill understood. Murid herpesvirus 4 (MuHV-4), a gammaherpesvirus of mice, is transmitted sexually. We show that this depends on the major virion envelope glycoprotein gp150. gp150 is redundant for host entry, and in vitro, it regulates rather than promotes cell binding. We show that gp150-deficient MuHV-4 reaches and replicates normally in the female genital tract after nasal infection but is poorly released from vaginal epithelial cells and fails to pass from the female to the male genital tract during sexual contact. Thus, we show that the regulation of virion binding is a key component of spontaneous gammaherpesvirus transmission.IMPORTANCE Gammaherpesviruses are responsible for many important diseases in both animals and humans. Some important aspects of their life cycle are still poorly understood. Key among these is viral transmission. Here we show that the major envelope glycoprotein of murid herpesvirus 4 functions not in entry or dissemination but in virion release to allow sexual transmission to new hosts.


Subject(s)
Glycoproteins/metabolism , Herpesviridae Infections/veterinary , Rhadinovirus/physiology , Sexually Transmitted Diseases, Viral/veterinary , Viral Envelope Proteins/metabolism , Virus Release , Animals , Disease Transmission, Infectious , Glycoproteins/genetics , Herpesviridae Infections/transmission , Herpesviridae Infections/virology , Sexually Transmitted Diseases, Viral/transmission , Sexually Transmitted Diseases, Viral/virology , Viral Envelope Proteins/genetics , Virus Attachment , Virus Internalization
12.
J Virol ; 91(21)2017 11 01.
Article in English | MEDLINE | ID: mdl-28794046

ABSTRACT

Virion transmembrane proteins (VTPs) mediate key functions in the herpesvirus infectious cycle. Cyprinid herpesvirus 3 (CyHV-3) is the archetype of fish alloherpesviruses. The present study was devoted to CyHV-3 VTPs. Using mass spectrometry approaches, we identified 16 VTPs of the CyHV-3 FL strain. Mutagenesis experiments demonstrated that eight of these proteins are essential for viral growth in vitro (open reading frame 32 [ORF32], ORF59, ORF81, ORF83, ORF99, ORF106, ORF115, and ORF131), and eight are nonessential (ORF25, ORF64, ORF65, ORF108, ORF132, ORF136, ORF148, and ORF149). Among the nonessential proteins, deletion of ORF25, ORF132, ORF136, ORF148, or ORF149 affects viral replication in vitro, and deletion of ORF25, ORF64, ORF108, ORF132, or ORF149 impacts plaque size. Lack of ORF148 or ORF25 causes attenuation in vivo to a minor or major extent, respectively. The safety and efficacy of a virus lacking ORF25 were compared to those of a previously described vaccine candidate deleted for ORF56 and ORF57 (Δ56-57). Using quantitative PCR, we demonstrated that the ORF25 deleted virus infects fish through skin infection and then spreads to internal organs as reported previously for the wild-type parental virus and the Δ56-57 virus. However, compared to the parental wild-type virus, the replication of the ORF25-deleted virus was reduced in intensity and duration to levels similar to those observed for the Δ56-57 virus. Vaccination of fish with a virus lacking ORF25 was safe but had low efficacy at the doses tested. This characterization of the virion transmembrane proteome of CyHV-3 provides a firm basis for further research on alloherpesvirus VTPs.IMPORTANCE Virion transmembrane proteins play key roles in the biology of herpesviruses. Cyprinid herpesvirus 3 (CyHV-3) is the archetype of fish alloherpesviruses and the causative agent of major economic losses in common and koi carp worldwide. In this study of the virion transmembrane proteome of CyHV-3, the major findings were: (i) the FL strain encodes 16 virion transmembrane proteins; (ii) eight of these proteins are essential for viral growth in vitro; (iii) seven of the nonessential proteins affect viral growth in vitro, and two affect virulence in vivo; and (iv) a mutant lacking ORF25 is highly attenuated but induces moderate immune protection. This study represents a major breakthrough in understanding the biology of CyHV-3 and will contribute to the development of prophylactic methods. It also provides a firm basis for the further research on alloherpesvirus virion transmembrane proteins.


Subject(s)
Herpesviridae Infections/metabolism , Membrane Proteins/metabolism , Proteome/analysis , Proteomics/methods , Viral Proteins/metabolism , Virion/metabolism , Virus Replication , Animals , Fishes/metabolism , Fishes/virology , Herpesviridae/metabolism , Herpesviridae/pathogenicity , Herpesviridae Infections/virology , Mass Spectrometry , Proteome/metabolism
13.
J Virol ; 90(4): 2039-51, 2016 02 15.
Article in English | MEDLINE | ID: mdl-26656682

ABSTRACT

UNLABELLED: Carbohydrates play major roles in host-virus interactions. It is therefore not surprising that, during coevolution with their hosts, viruses have developed sophisticated mechanisms to hijack for their profit different pathways of glycan synthesis. Thus, the Bo17 gene of Bovine herpesvirus 4 (BoHV-4) encodes a homologue of the cellular core 2 protein ß-1,6-N-acetylglucosaminyltransferase-mucin type (C2GnT-M), which is a key player for the synthesis of complex O-glycans. Surprisingly, we show in this study that, as opposed to what is observed for the cellular enzyme, two different mRNAs are encoded by the Bo17 gene of all available BoHV-4 strains. While the first one corresponds to the entire coding sequence of the Bo17 gene, the second results from the splicing of a 138-bp intron encoding critical residues of the enzyme. Antibodies generated against the Bo17 C terminus showed that the two forms of Bo17 are expressed in BoHV-4 infected cells, but enzymatic assays revealed that the spliced form is not active. In order to reveal the function of these two forms, we then generated recombinant strains expressing only the long or the short form of Bo17. Although we did not highlight replication differences between these strains, glycomic analyses and lectin neutralization assays confirmed that the splicing of the Bo17 gene gives the potential to BoHV-4 to fine-tune the global level of core 2 branching activity in the infected cell. Altogether, these results suggest the existence of new mechanisms to regulate the activity of glycosyltransferases from the Golgi apparatus. IMPORTANCE: Viruses are masters of adaptation that hijack cellular pathways to allow their growth. Glycans play a central role in many biological processes, and several studies have highlighted mechanisms by which viruses can affect glycosylation. Glycan synthesis is a nontemplate process regulated by the availability of key glycosyltransferases. Interestingly, bovine herpesvirus 4 encodes one such enzyme which is a key enzyme for the synthesis of complex O-glycans. In this study, we show that, in contrast to cellular homologues, this virus has evolved to alternatively express two proteins from this gene. While the first one is enzymatically active, the second results from the alternative splicing of the region encoding the catalytic site of the enzyme. We postulate that this regulatory mechanism could allow the virus to modulate the synthesis of some particular glycans for function at the location and/or the moment of infection.


Subject(s)
Alternative Splicing , Gene Expression Regulation, Viral , Herpesvirus 4, Bovine/enzymology , Herpesvirus 4, Bovine/genetics , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Animals , Cattle , Cells, Cultured , Gene Expression Profiling
14.
J Virol ; 90(5): 2455-72, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26676769

ABSTRACT

UNLABELLED: Gammaherpesviruses are important human and animal pathogens. Despite the fact that they display the classical architecture of herpesviruses, the function of most of their structural proteins is still poorly defined. This is especially true for tegument proteins. Interestingly, a potential role in immune evasion has recently been proposed for the tegument protein encoded by Kaposi's sarcoma-associated herpesvirus open reading frame 63 (ORF63). To gain insight about the roles of ORF63 in the life cycle of a gammaherpesvirus, we generated null mutations in the ORF63 gene of murid herpesvirus 4 (MuHV-4). We showed that disruption of ORF63 was associated with a severe MuHV-4 growth deficit both in vitro and in vivo. The latter deficit was mainly associated with a defect of replication in the lung but did not affect the establishment of latency in the spleen. From a functional point of view, inhibition of caspase-1 or the inflammasome did not restore the growth of the ORF63-deficient mutant, suggesting that the observed deficit was not associated with the immune evasion mechanism identified previously. Moreover, this growth deficit was also not associated with a defect in virion egress from the infected cells. In contrast, it appeared that MuHV-4 ORF63-deficient mutants failed to address most of their capsids to the nucleus during entry into the host cell, suggesting that ORF63 plays a role in capsid movement. In the future, ORF63 could therefore be considered a target to block gammaherpesvirus infection at a very early stage of the infection. IMPORTANCE: The important diseases caused by gammaherpesviruses in human and animal populations justify a better understanding of their life cycle. In particular, the role of most of their tegument proteins is still largely unknown. In this study, we used murid herpesvirus 4, a gammaherpesvirus infecting mice, to decipher the role of the protein encoded by the viral ORF63 gene. We showed that the absence of this protein is associated with a severe growth deficit both in vitro and in vivo that was mainly due to impaired migration of viral capsids toward the nucleus during entry. Together, our results provide new insights about the life cycle of gammaherpesviruses and could allow the development of new antiviral strategies aimed at blocking gammaherpesvirus infection at the very early stages.


Subject(s)
Biological Transport , Capsid/metabolism , Rhadinovirus/physiology , Viral Proteins/metabolism , Virus Internalization , Animals , Cell Line , Cricetinae , Female , Gene Deletion , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Histocytochemistry , Lung/pathology , Lung/virology , Mice, Inbred BALB C , Rhadinovirus/genetics , Rhadinovirus/growth & development , Viral Proteins/genetics
15.
Reprod Fertil Dev ; 28(5): 628-39, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25482458

ABSTRACT

When derived from chicken embryos, avian primordial germ cells (PGCs) have been reported to keep their germline-specific properties and proliferative potential even after long-term culture and genetic modifications. Few teams to date have reported such long-term expansion and engineering without differentiation of primary avian PGCs' cultures. We have developed original and robust methods that allow more than 1 year culture, expansion and cryobanking of primary cultures of PGCs without obvious effects on their biological properties, including their ability to colonise the genital ridges. Overall, 38% of embryonic samples gave rise to PGCs lines derived from three commercial layers and two Belgian endangered breeds. The lines kept their proliferative potential and their characteristic PGCs phenotype after 20 months in culture, whether or not interrupted by a cryopreservation step. All the resulting lines appeared devoid of female cells, although initially pooled from male and female embryos. Labelled PGCs from 12 long-term cultured lines colonised the genital ridges of recipient embryos. Thus, this procedure allows derivation, long-term expansion and cryobanking of primary cultures of PGCs without obvious changes to their original characteristics, providing an alternative access to applications in avian biotechnology and preservation of genetic resources.


Subject(s)
Cell Movement , Cell Proliferation , Chickens/physiology , Cryopreservation/veterinary , Endangered Species , Germ Cells/physiology , Gonads/embryology , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Chick Embryo , Chickens/genetics , Female , Germ Cells/metabolism , Germ Cells/transplantation , Male , Phenotype , Sex Determination Analysis/veterinary , Time Factors
16.
PLoS Pathog ; 9(10): e1003753, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24204281

ABSTRACT

Human gammaherpesviruses are associated with the development of lymphomas and epithelial malignancies. The heterogeneity of these tumors reflects the ability of these viruses to route infection to different cell types at various stages of their lifecycle. While the Epstein Barr virus uses gp42--human leukocyte antigen class II interaction as a switch of cell tropism, the molecular mechanism that orientates tropism of rhadinoviruses is still poorly defined. Here, we used bovine herpesvirus 4 (BoHV-4) to further elucidate how rhadinoviruses regulate their infectivity. In the absence of any gp42 homolog, BoHV-4 exploits the alternative splicing of its Bo10 gene to produce distinct viral populations that behave differently based on the originating cell. While epithelial cells produce virions with high levels of the accessory envelope protein gp180, encoded by a Bo10 spliced product, myeloid cells express reduced levels of gp180. As a consequence, virions grown in epithelial cells are hardly infectious for CD14+ circulating cells, but are relatively resistant to antibody neutralization due to the shielding property of gp180 for vulnerable entry epitopes. In contrast, myeloid virions readily infect CD14+ circulating cells but are easily neutralized. This molecular switch could therefore allow BoHV-4 to promote either, on the one hand, its dissemination into the organism, or, on the other hand, its transmission between hosts.


Subject(s)
Alternative Splicing/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Herpesvirus 4, Bovine/immunology , Viral Proteins/immunology , Alternative Splicing/genetics , Animals , Cattle , Cell Line , Herpesvirus 4, Bovine/genetics , Humans , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/immunology , Rabbits
17.
PLoS Pathog ; 9(4): e1003292, 2013.
Article in English | MEDLINE | ID: mdl-23593002

ABSTRACT

Transmission is a matter of life or death for pathogen lineages and can therefore be considered as the main motor of their evolution. Gammaherpesviruses are archetypal pathogenic persistent viruses which have evolved to be transmitted in presence of specific immune response. Identifying their mode of transmission and their mechanisms of immune evasion is therefore essential to develop prophylactic and therapeutic strategies against these infections. As the known human gammaherpesviruses, Epstein-Barr virus and Kaposi's Sarcoma-associated Herpesvirus are host-specific and lack a convenient in vivo infection model; related animal gammaherpesviruses, such as murine gammaherpesvirus-68 (MHV-68), are commonly used as general models of gammaherpesvirus infections in vivo. To date, it has however never been possible to monitor viral excretion or virus transmission of MHV-68 in laboratory mice population. In this study, we have used MHV-68 associated with global luciferase imaging to investigate potential excretion sites of this virus in laboratory mice. This allowed us to identify a genital excretion site of MHV-68 following intranasal infection and latency establishment in female mice. This excretion occurred at the external border of the vagina and was dependent on the presence of estrogens. However, MHV-68 vaginal excretion was not associated with vertical transmission to the litter or with horizontal transmission to female mice. In contrast, we observed efficient virus transmission to naïve males after sexual contact. In vivo imaging allowed us to show that MHV-68 firstly replicated in penis epithelium and corpus cavernosum before spreading to draining lymph nodes and spleen. All together, those results revealed the first experimental transmission model for MHV-68 in laboratory mice. In the future, this model could help us to better understand the biology of gammaherpesviruses and could also allow the development of strategies that could prevent the spread of these viruses in natural populations.


Subject(s)
Gammaherpesvirinae/pathogenicity , Genitalia, Female/virology , Genitalia, Male/virology , Herpesviridae Infections/transmission , Sexually Transmitted Diseases, Viral/virology , Animals , Cell Line , Cricetinae , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Immune Evasion , Infectious Disease Transmission, Vertical , Male , Mice , Mice, Inbred BALB C , Models, Animal , Virus Replication , Virus Shedding
18.
J Virol ; 87(19): 10828-42, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23903840

ABSTRACT

Glycoprotein B (gB) is a conserved herpesvirus virion component implicated in membrane fusion. As with many-but not all-herpesviruses, the gB of murid herpesvirus 4 (MuHV-4) is cleaved into disulfide-linked subunits, apparently by furin. Preventing gB cleavage for some herpesviruses causes minor infection deficits in vitro, but what the cleavage contributes to host colonization has been unclear. To address this, we mutated the furin cleavage site (R-R-K-R) of the MuHV-4 gB. Abolishing gB cleavage did not affect its expression levels, glycosylation, or antigenic conformation. In vitro, mutant viruses entered fibroblasts and epithelial cells normally but had a significant entry deficit in myeloid cells such as macrophages and bone marrow-derived dendritic cells. The deficit in myeloid cells was not due to reduced virion binding or endocytosis, suggesting that gB cleavage promotes infection at a postendocytic entry step, presumably viral membrane fusion. In vivo, viruses lacking gB cleavage showed reduced lytic spread in the lungs. Alveolar epithelial cell infection was normal, but alveolar macrophage infection was significantly reduced. Normal long-term latency in lymphoid tissue was established nonetheless.


Subject(s)
Glycoproteins/metabolism , Lung/virology , Myeloid Cells/virology , Rhadinovirus/physiology , Viral Envelope Proteins/metabolism , Animals , Antibodies, Neutralizing/immunology , Base Sequence , Blotting, Western , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/virology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Flow Cytometry , Fluorescent Antibody Technique , Furin/metabolism , Glycoproteins/genetics , Lung/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/virology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Viral Envelope Proteins/genetics , Virion , Virus Replication
19.
PLoS Pathog ; 8(9): e1002935, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028329

ABSTRACT

Gamma-herpesviruses persist in lymphocytes and cause disease by driving their proliferation. Lymphocyte infection is therefore a key pathogenetic event. Murid Herpesvirus-4 (MuHV-4) is a rhadinovirus that like the related Kaposi's Sarcoma-associated Herpesvirus persists in B cells in vivo yet infects them poorly in vitro. Here we used MuHV-4 to understand how virion tropism sets the path to lymphocyte colonization. Virions that were highly infectious in vivo showed a severe post-binding block to B cell infection. Host entry was accordingly an epithelial infection and B cell infection a secondary event. Macrophage infection by cell-free virions was also poor, but improved markedly when virion binding improved or when macrophages were co-cultured with infected fibroblasts. Under the same conditions B cell infection remained poor; it improved only when virions came from macrophages. This reflected better cell penetration and correlated with antigenic changes in the virion fusion complex. Macrophages were seen to contact acutely infected epithelial cells, and cre/lox-based virus tagging showed that almost all the virus recovered from lymphoid tissue had passed through lysM(+) and CD11c(+) myeloid cells. Thus MuHV-4 reached B cells in 3 distinct stages: incoming virions infected epithelial cells; infection then passed to myeloid cells; glycoprotein changes then allowed B cell infection. These data identify new complexity in rhadinovirus infection and potentially also new vulnerability to intervention.


Subject(s)
B-Lymphocytes/virology , Epithelial Cells/virology , Herpesviridae Infections/virology , Macrophages/virology , Rhadinovirus/physiology , Rhadinovirus/pathogenicity , Tumor Virus Infections/virology , 3T3 Cells , Animals , CD11c Antigen , Cell Line , Cricetinae , Fibroblasts/virology , HEK293 Cells , Herpesviridae Infections/immunology , Humans , Mice , Mice, Inbred C57BL , Myeloid Cells/virology , Rhadinovirus/immunology , Tumor Virus Infections/immunology , Viral Envelope Proteins/metabolism , Virus Internalization
20.
ACS Nanosci Au ; 4(2): 136-145, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38644967

ABSTRACT

The SARS-CoV-2 pandemic spurred numerous research endeavors to comprehend the virus and mitigate its global severity. Understanding the binding interface between the virus and human receptors is pivotal to these efforts and paramount to curbing infection and transmission. Here we employ atomic force microscopy and steered molecular dynamics simulation to explore SARS-CoV-2 receptor binding domain (RBD) variants and angiotensin-converting enzyme 2 (ACE2), examining the impact of mutations at key residues upon binding affinity. Our results show that the Omicron and Delta variants possess strengthened binding affinity in comparison to the Mu variant. Further, using sera from individuals either vaccinated or with acquired immunity following Delta strain infection, we assess the impact of immunity upon variant RBD/ACE2 complex formation. Single-molecule force spectroscopy analysis suggests that vaccination before infection may provide stronger protection across variants. These results underscore the need to monitor antigenic changes in order to continue developing innovative and effective SARS-CoV-2 abrogation strategies.

SELECTION OF CITATIONS
SEARCH DETAIL