Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Nat Immunol ; 22(1): 25-31, 2021 01.
Article in English | MEDLINE | ID: mdl-33154590

ABSTRACT

Clinical manifestations of COVID-19 caused by the new coronavirus SARS-CoV-2 are associated with age1,2. Adults develop respiratory symptoms, which can progress to acute respiratory distress syndrome (ARDS) in the most severe form, while children are largely spared from respiratory illness but can develop a life-threatening multisystem inflammatory syndrome (MIS-C)3-5. Here, we show distinct antibody responses in children and adults after SARS-CoV-2 infection. Adult COVID-19 cohorts had anti-spike (S) IgG, IgM and IgA antibodies, as well as anti-nucleocapsid (N) IgG antibody, while children with and without MIS-C had reduced breadth of anti-SARS-CoV-2-specific antibodies, predominantly generating IgG antibodies specific for the S protein but not the N protein. Moreover, children with and without MIS-C had reduced neutralizing activity as compared to both adult COVID-19 cohorts, indicating a reduced protective serological response. These results suggest a distinct infection course and immune response in children independent of whether they develop MIS-C, with implications for developing age-targeted strategies for testing and protecting the population.


Subject(s)
Antibodies, Viral/immunology , Antibody Formation/immunology , COVID-19/immunology , Nucleocapsid Proteins/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Adolescent , Adult , Aged , COVID-19/virology , Child , Child, Preschool , Female , Humans , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Male , Middle Aged , SARS-CoV-2/physiology , Young Adult
2.
Cell Mol Life Sci ; 81(1): 71, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38300320

ABSTRACT

Hexosylceramides (HexCer) are implicated in the infection process of various pathogens. However, the molecular and cellular functions of HexCer in infectious cycles are poorly understood. Investigating the enveloped virus Uukuniemi (UUKV), a bunyavirus of the Phenuiviridae family, we performed a lipidomic analysis with mass spectrometry and determined the lipidome of both infected cells and derived virions. We found that UUKV alters the processing of HexCer to glycosphingolipids (GSL) in infected cells. The infection resulted in the overexpression of glucosylceramide (GlcCer) synthase (UGCG) and the specific accumulation of GlcCer and its subsequent incorporation into viral progeny. UUKV and several pathogenic bunyaviruses relied on GlcCer in the viral envelope for binding to various host cell types. Overall, our results indicate that GlcCer is a structural determinant of virions crucial for bunyavirus infectivity. This study also highlights the importance of glycolipids on virions in facilitating interactions with host cell receptors and infectious entry of enveloped viruses.


Subject(s)
Orthobunyavirus , Glucosylceramides , Virus Attachment , Lipidomics , Mass Spectrometry
3.
J Biol Chem ; 296: 100111, 2021.
Article in English | MEDLINE | ID: mdl-33229438

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a ß-coronavirus, is the causative agent of the COVID-19 pandemic. Like for other coronaviruses, its particles are composed of four structural proteins: spike (S), envelope (E), membrane (M), and nucleoprotein (N) proteins. The involvement of each of these proteins and their interactions are critical for assembly and production of ß-coronavirus particles. Here, we sought to characterize the interplay of SARS-CoV-2 structural proteins during the viral assembly process. By combining biochemical and imaging assays in infected versus transfected cells, we show that E and M regulate intracellular trafficking of S as well as its intracellular processing. Indeed, the imaging data reveal that S is relocalized at endoplasmic reticulum (ER)-Golgi intermediate compartment (ERGIC) or Golgi compartments upon coexpression of E or M, as observed in SARS-CoV-2-infected cells, which prevents syncytia formation. We show that a C-terminal retrieval motif in the cytoplasmic tail of S is required for its M-mediated retention in the ERGIC, whereas E induces S retention by modulating the cell secretory pathway. We also highlight that E and M induce a specific maturation of N-glycosylation of S, independently of the regulation of its localization, with a profile that is observed both in infected cells and in purified viral particles. Finally, we show that E, M, and N are required for optimal production of virus-like-particles. Altogether, these results highlight how E and M proteins may influence the properties of S proteins and promote the assembly of SARS-CoV-2 viral particles.


Subject(s)
Coronavirus Envelope Proteins/genetics , Nucleocapsid Proteins/genetics , SARS-CoV-2/growth & development , Spike Glycoprotein, Coronavirus/genetics , Viral Matrix Proteins/genetics , Virion/growth & development , Virus Assembly/physiology , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Coronavirus Envelope Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum/virology , Gene Expression , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Golgi Apparatus/virology , HEK293 Cells , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Hepatocytes/virology , Host-Pathogen Interactions/genetics , Humans , Nucleocapsid Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Vero Cells , Viral Matrix Proteins/metabolism , Virion/genetics , Virion/metabolism , Virus Internalization , Virus Release/physiology
4.
Kidney Int ; 101(2): 390-402, 2022 02.
Article in English | MEDLINE | ID: mdl-34856313

ABSTRACT

The level of protection achieved by the standard two doses of COVID-19 mRNA vaccines in patients receiving maintenance hemodialysis (MHD) remains unclear. To study this we used the French Renal Epidemiology and Information Network (REIN) Registry to compare the incidence and severity of 1474 cases of COVID-19 diagnosed in patients receiving MHD after none, one or two doses of vaccine. Vaccination significantly reduce COVID-19 incidence and severity, but 11% of patients infected after two doses still died. Lack of vaccinal protection in patients naïve for SARS-CoV-2 could be due to defective Tfh response [38% of patients with negative spike-specific CD4+ T-cell interferon gamma release assay] and failure to generate viral neutralizing titers of anti-spike receptor binding domain (RBD) IgGs (63% of patients with titer at or under 997 BAU/ml, defining low/no responders) after two doses of vaccine. To improve protection, a third dose of vaccine was administered to 75 patients [57 low/no responders, 18 high responders after two doses] from the ROMANOV cohort that prospectively enrolled patients receiving MHD vaccinated with BNT162b2 (Pfizer). Tolerance to the third dose was excellent. High responders to two doses did not generate more anti-RBD IgGs after three doses but had more side effects. Importantly, 31 (54%) of low/no responders to two doses reached neutralizing titers of anti-RBD IgGs after three doses. A positive interferon gamma release assay and/or suboptimal titer of anti-RBD IgGs after two doses were the only predictive variables for response to three doses in multivariate analysis. Thus, the standard scheme of vaccination insufficiently protects patients receiving MHD. Anti-RBD IgG and specific CD4+ T-cell response after two doses can guide personalized administration of the third dose, which improves the humoral response of SARS-CoV-2-naïve patients receiving MHD.


Subject(s)
BNT162 Vaccine , COVID-19 , Antibodies, Viral , Humans , Renal Dialysis/adverse effects , SARS-CoV-2 , Vaccines, Synthetic , mRNA Vaccines
5.
Am J Transplant ; 22(5): 1442-1450, 2022 05.
Article in English | MEDLINE | ID: mdl-35114060

ABSTRACT

Kidney transplant recipients (KTRs) have reduced ability to mount adequate antibody response after two doses of the COVID-19 mRNA vaccine. French health authorities have allowed a third booster dose (D3) for KTRs, but their response is heterogeneous and tools able to discriminate the responders are lacking. Anti-RBD IgG titers (chemiluminescence immunoassay), spike-specific cellular responses (IFN-γ-releasing assay, IGRA), and in vitro serum neutralization of the virus (the best available correlate of protection), were evaluated 7-14 days after the second dose (D2) of BNT162b2 vaccine in 93 KTRs. Among the 73 KTRs, whose serum did not neutralize SARS-CoV-2 in vitro after D2, 14 (19%) acquired this capacity after D3, and were considered as "responders." Exploratory univariate analysis identified short time from transplantation and high maintenance immunosuppression as detrimental factors for the response to D3. In addition, any of the presence of anti-RBD IgGs and/or positive IGRA after D2 was predictive of response to D3. By contrast, none of the KTRs with both a negative serology and IGRA responded to D3. In summary, routinely available bioassays performed after D2 allow identifying KTRs that will respond to a booster D3. These results pave the way for the personalization of vaccination strategy in KTRs.


Subject(s)
COVID-19 , Kidney Transplantation , Antibodies, Neutralizing , Antibodies, Viral , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines , Humans , SARS-CoV-2 , Vaccines, Synthetic , mRNA Vaccines
6.
Emerg Infect Dis ; 26(1): 104-113, 2020 01.
Article in English | MEDLINE | ID: mdl-31855143

ABSTRACT

We conducted an in-depth characterization of the Nipah virus (NiV) isolate previously obtained from a Pteropus lylei bat in Cambodia in 2003 (CSUR381). We performed full-genome sequencing and phylogenetic analyses and confirmed CSUR381 is part of the NiV-Malaysia genotype. In vitro studies revealed similar cell permissiveness and replication of CSUR381 (compared with 2 other NiV isolates) in both bat and human cell lines. Sequence alignments indicated conservation of the ephrin-B2 and ephrin-B3 receptor binding sites, the glycosylation site on the G attachment protein, as well as the editing site in phosphoprotein, suggesting production of nonstructural proteins V and W, known to counteract the host innate immunity. In the hamster animal model, CSUR381 induced lethal infections. Altogether, these data suggest that the Cambodia bat-derived NiV isolate has high pathogenic potential and, thus, provide insight for further studies and better risk assessment for future NiV outbreaks in Southeast Asia.


Subject(s)
Chiroptera/virology , Henipavirus Infections/veterinary , Nipah Virus/pathogenicity , Animals , Cambodia , Genome, Viral/genetics , Henipavirus Infections/epidemiology , Henipavirus Infections/virology , Humans , Nipah Virus/genetics , Phylogeny , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Whole Genome Sequencing
7.
J Virol ; 93(13)2019 07 01.
Article in English | MEDLINE | ID: mdl-31019048

ABSTRACT

Fatal neurological syndromes can occur after measles virus (MeV) infection of the brain. The mechanisms controlling MeV spread within the central nervous system (CNS) remain poorly understood. We analyzed the role of type I interferon (IFN-I) receptor (IFNAR) signaling in the control of MeV infection in a murine model of brain infection. Using organotypic brain cultures (OBC) from wild-type and IFNAR-knockout (IFNARKO) transgenic mice ubiquitously expressing the human SLAM (CD150) receptor, the heterogeneity of the permissiveness of different CNS cell types to MeV infection was characterized. In the absence of IFNAR signaling, MeV propagated significantly better in explant slices. In OBC from IFNAR-competent mice, while astrocytes and microglia were infected on the day of explant preparation, they became refractory to infection with time, in contrast to neurons and oligodendrocytes, which remained permissive to infection. This selective loss of permissiveness to MeV infection was not observed in IFNARKO mouse OBC. Accordingly, the development of astrogliosis related to the OBC procedure was exacerbated in the presence of IFNAR signaling. In the hippocampus, this astrogliosis was characterized by a change in the astrocyte phenotype and by an increase of IFN-I transcripts. A proteome analysis showed the upregulation of 84 out of 111 secreted proteins. In the absence of IFNAR, only 27 secreted proteins were upregulated, and none of these were associated with antiviral activities. Our results highlight the essential role of the IFN-I response in astrogliosis and in the permissiveness of astrocytes and microglia that could control MeV propagation throughout the CNS.IMPORTANCE Measles virus (MeV) can infect the central nervous system (CNS), with dramatic consequences. The mechanisms controlling MeV invasion of the CNS remain ill-defined since most previous data were obtained from postmortem analysis. Here, we highlight for the first time the crucial role of the type I interferon (IFN-I) response not only in the control of CNS invasion but also in the early permissiveness of glial cells to measles virus infection.


Subject(s)
Astrocytes/virology , Measles virus/metabolism , Measles/metabolism , Microglia/virology , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/physiology , Animals , Antiviral Agents/pharmacology , Astrocytes/pathology , Brain/virology , Central Nervous System/virology , Cytokines , Female , Hippocampus/pathology , Hippocampus/virology , Humans , Male , Measles/pathology , Measles/virology , Mice , Mice, Knockout , Neurons/virology , Oligodendroglia/virology , Receptor, Interferon alpha-beta/genetics , Signal Transduction/genetics , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism
8.
J Virol ; 93(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30487282

ABSTRACT

During a measles virus (MeV) epidemic in 2009 in South Africa, measles inclusion body encephalitis (MIBE) was identified in several HIV-infected patients. Years later, children are presenting with subacute sclerosing panencephalitis (SSPE). To investigate the features of established MeV neuronal infections, viral sequences were analyzed from brain tissue samples of a single SSPE case and compared with MIBE sequences previously obtained from patients infected during the same epidemic. Both the SSPE and the MIBE viruses had amino acid substitutions in the ectodomain of the F protein that confer enhanced fusion properties. Functional analysis of the fusion complexes confirmed that both MIBE and SSPE F protein mutations promoted fusion with less dependence on interaction by the viral receptor-binding protein with known MeV receptors. While the SSPE F required the presence of a homotypic attachment protein, MeV H, in order to fuse, MIBE F did not. Both F proteins had decreased thermal stability compared to that of the corresponding wild-type F protein. Finally, recombinant viruses expressing MIBE or SSPE fusion complexes spread in the absence of known MeV receptors, with MIBE F-bearing viruses causing large syncytia in these cells. Our results suggest that alterations to the MeV fusion complex that promote fusion and cell-to-cell spread in the absence of known MeV receptors is a key property for infection of the brain.IMPORTANCE Measles virus can invade the central nervous system (CNS) and cause severe neurological complications, such as MIBE and SSPE. However, mechanisms by which MeV enters the CNS and triggers the disease remain unclear. We analyzed viruses from brain tissue of individuals with MIBE or SSPE, infected during the same epidemic, after the onset of neurological disease. Our findings indicate that the emergence of hyperfusogenic MeV F proteins is associated with infection of the brain. We also demonstrate that hyperfusogenic F proteins permit MeV to enter cells and spread without the need to engage nectin-4 or CD150, known receptors for MeV that are not present on neural cells.


Subject(s)
Measles virus/genetics , Subacute Sclerosing Panencephalitis/genetics , Viral Fusion Proteins/genetics , Amino Acid Substitution , Animals , Brain/virology , Cell Adhesion Molecules/metabolism , Chlorocebus aethiops , Epidemics , Female , Genotype , Giant Cells/virology , HEK293 Cells , Humans , Male , Measles/epidemiology , Measles/metabolism , Measles/virology , Mutation , Neurons/virology , South Africa , Subacute Sclerosing Panencephalitis/virology , Vero Cells , Viral Fusion Proteins/metabolism
9.
J Virol ; 93(8)2019 04 15.
Article in English | MEDLINE | ID: mdl-30728259

ABSTRACT

A clinical isolate of measles virus (MeV) bearing a single amino acid alteration in the viral fusion protein (F; L454W) was previously identified in two patients with lethal sequelae of MeV central nervous system (CNS) infection. The mutation dysregulated the viral fusion machinery so that the mutated F protein mediated cell fusion in the absence of known MeV cellular receptors. While this virus could feasibly have arisen via intrahost evolution of the wild-type (wt) virus, it was recently shown that the same mutation emerged under the selective pressure of small-molecule antiviral treatment. Under these conditions, a potentially neuropathogenic variant emerged outside the CNS. While CNS adaptation of MeV was thought to generate viruses that are less fit for interhost spread, we show that two animal models can be readily infected with CNS-adapted MeV via the respiratory route. Despite bearing a fusion protein that is less stable at 37°C than the wt MeV F, this virus infects and replicates in cotton rat lung tissue more efficiently than the wt virus and is lethal in a suckling mouse model of MeV encephalitis even with a lower inoculum. Thus, either during lethal MeV CNS infection or during antiviral treatment in vitro, neuropathogenic MeV can emerge, can infect new hosts via the respiratory route, and is more pathogenic (at least in these animal models) than wt MeV.IMPORTANCE Measles virus (MeV) infection can be severe in immunocompromised individuals and lead to complications, including measles inclusion body encephalitis (MIBE). In some cases, MeV persistence and subacute sclerosing panencephalitis (SSPE) occur even in the face of an intact immune response. While they are relatively rare complications of MeV infection, MIBE and SSPE are lethal. This work addresses the hypothesis that despite a dysregulated viral fusion complex, central nervous system (CNS)-adapted measles virus can spread outside the CNS within an infected host.


Subject(s)
Central Nervous System/virology , Encephalitis, Viral , Inclusion Bodies, Viral , Lung/virology , Measles virus/physiology , Measles , Mutation, Missense , Viral Fusion Proteins , Virus Replication , Amino Acid Substitution , Animals , Central Nervous System/metabolism , Chlorocebus aethiops , Disease Models, Animal , Encephalitis, Viral/genetics , Encephalitis, Viral/metabolism , Encephalitis, Viral/transmission , Humans , Inclusion Bodies, Viral/genetics , Inclusion Bodies, Viral/metabolism , Lung/metabolism , Measles/metabolism , Measles/transmission , Mice , Mice, Transgenic , Sigmodontinae , Vero Cells , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism
10.
J Infect Dis ; 218(2): 218-227, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29566184

ABSTRACT

Background: The emerging zoonotic paramyxovirus Nipah virus (NiV) causes severe respiratory and neurological disease in humans, with high fatality rates. Nipah virus can be transmitted via person-to-person contact, posing a high risk for epidemic outbreaks. However, a broadly applicable approach for human NiV outbreaks in field settings is lacking. Methods: We engineered new antiviral lipopeptides and analyzed in vitro fusion inhibition to identify an optimal candidate for prophylaxis of NiV infection in the lower respiratory tract, and we assessed antiviral efficiency in 2 different animal models. Results: We show that lethal NiV infection can be prevented with lipopeptides delivered via the respiratory route in both hamsters and nonhuman primates. By targeting retention of peptides for NiV prophylaxis in the respiratory tract, we avoid its systemic delivery in individuals who need only prevention, and thus we increase the safety of treatment and enhance utility of the intervention. Conclusions: The experiments provide a proof of concept for the use of antifusion lipopeptides for prophylaxis of lethal NiV. These results advance the goal of rational development of potent lipopeptide inhibitors with desirable pharmacokinetic and biodistribution properties and a safe effective delivery method to target NiV and other pathogenic viruses.


Subject(s)
Chemoprevention/methods , Henipavirus Infections/prevention & control , Lipopeptides/administration & dosage , Nipah Virus/physiology , Primate Diseases/prevention & control , Viral Envelope Proteins/antagonists & inhibitors , Viral Fusion Protein Inhibitors/administration & dosage , Animals , Bronchopneumonia/prevention & control , Bronchopneumonia/veterinary , Chlorocebus aethiops , Disease Models, Animal , Female , Humans , Male , Mesocricetus
11.
J Virol ; 90(15): 6642-6656, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27170753

ABSTRACT

UNLABELLED: Nonsegmented negative-stranded RNA viruses, or members of the order Mononegavirales, share a conserved gene order and the use of elaborate transcription and replication machinery made up of at least four molecular partners. These partners have coevolved with the acquisition of the permanent encapsidation of the entire genome by the nucleoprotein (N) and the use of this N-RNA complex as a template for the viral polymerase composed of the phosphoprotein (P) and the large enzymatic protein (L). Not only is P required for polymerase function, but it also stabilizes the L protein through an unknown underlying molecular mechanism. By using NVP-AUY922 and/or 17-dimethylaminoethylamino-17-demethoxygeldanamycin as specific inhibitors of cellular heat shock protein 90 (HSP90), we found that efficient chaperoning of L by HSP90 requires P in the measles, Nipah, and vesicular stomatitis viruses. While the production of P remains unchanged in the presence of HSP90 inhibitors, the production of soluble and functional L requires both P and HSP90 activity. Measles virus P can bind the N terminus of L in the absence of HSP90 activity. Both HSP90 and P are required for the folding of L, as evidenced by a luciferase reporter insert fused within measles virus L. HSP90 acts as a true chaperon; its activity is transient and dispensable for the activity of measles and Nipah virus polymerases of virion origin. That the cellular chaperoning of a viral polymerase into a soluble functional enzyme requires the assistance of another viral protein constitutes a new paradigm that seems to be conserved within the Mononegavirales order. IMPORTANCE: Viruses are obligate intracellular parasites that require a cellular environment for their replication. Some viruses particularly depend on the cellular chaperoning apparatus. We report here that for measles virus, successful chaperoning of the viral L polymerase mediated by heat shock protein 90 (HSP90) requires the presence of the viral phosphoprotein (P). Indeed, while P protein binds to the N terminus of L independently of HSP90 activity, both HSP90 and P are required to produce stable, soluble, folded, and functional L proteins. Once formed, the mature P+L complex no longer requires HSP90 to exert its polymerase functions. Such a new paradigm for the maturation of a viral polymerase appears to be conserved in several members of the Mononegavirales order, including the Nipah and vesicular stomatitis viruses.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , HSP90 Heat-Shock Proteins/metabolism , Henipavirus Infections/metabolism , Measles/metabolism , Phosphoproteins/metabolism , Protein Folding , Animals , Chlorocebus aethiops , HSP90 Heat-Shock Proteins/chemistry , HeLa Cells , Henipavirus Infections/virology , Humans , Measles/virology , Measles virus/physiology , Mice , Nipah Virus/physiology , Nucleoproteins/metabolism , Protein Binding , Rhabdoviridae Infections/metabolism , Rhabdoviridae Infections/virology , Vero Cells , Vesiculovirus/physiology , Viral Proteins/metabolism , Virion/physiology , Virus Replication
12.
J Virol ; 87(24): 13785-94, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24109233

ABSTRACT

Measles virus (MV) infection causes an acute childhood disease that can include infection of the central nervous system and can rarely progress to severe neurological disease for which there is no specific treatment. We generated potent antiviral peptide inhibitors of MV entry and spreading and MV-induced cell fusion. Dimers of MV-specific peptides derived from the C-terminal heptad repeat region of the MV fusion protein, conjugated to cholesterol, efficiently protect SLAM transgenic mice from fatal MV infection. Fusion inhibitors hold promise for the prophylaxis of MV infection in unvaccinated and immunocompromised people, as well as potential for the treatment of grave neurological complications of measles.


Subject(s)
Antiviral Agents/pharmacology , Brain/virology , Measles virus/drug effects , Measles/prevention & control , Viral Fusion Proteins/antagonists & inhibitors , Animals , Brain/drug effects , Cell Line , Humans , Measles/drug therapy , Measles/mortality , Measles/virology , Measles virus/genetics , Measles virus/physiology , Mice , Mice, Transgenic , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virus Internalization/drug effects
13.
J Infect Dis ; 207(1): 142-51, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23089589

ABSTRACT

Hendra virus (HeV) and Nipah virus (NiV) are closely related, recently emerged paramyxoviruses that form Henipavirus genus and are capable of causing considerable morbidity and mortality in a number of mammalian species, including humans. However, in contrast to many other species and despite expression of functional virus entry receptors, mice are resistant to henipavirus infection. We report here the susceptibility of mice deleted for the type I interferon receptor (IFNAR-KO) to both HeV and NiV. Intraperitoneally infected mice developed fatal encephalitis, with pathology and immunohistochemical features similar to what was found in humans. Viral RNA was found in the majority of analyzed organs, and sublethally infected animals developed virus-specific neutralizing antibodies. Altogether, these results reveal IFNAR-KO mice as a new small animal model to study HeV and NiV pathogenesis, prophylaxis, and treatment and suggest the critical role of type I interferon signaling in the control of henipavirus infection.


Subject(s)
Antibodies, Viral/immunology , Encephalitis, Viral/prevention & control , Henipavirus Infections/prevention & control , Henipavirus/immunology , Interferon Type I/genetics , Animals , Antibodies, Neutralizing , Antibody Specificity , Brain/virology , Cells, Cultured , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/mortality , Encephalitis, Viral/virology , Hendra Virus/genetics , Hendra Virus/immunology , Hendra Virus/pathogenicity , Henipavirus/genetics , Henipavirus/pathogenicity , Henipavirus Infections/immunology , Henipavirus Infections/mortality , Henipavirus Infections/virology , Humans , Interferon Type I/immunology , Mice , Mice, Knockout , Neuroglia/virology , Nipah Virus/genetics , Nipah Virus/immunology , Nipah Virus/pathogenicity , RNA, Viral/analysis , Signal Transduction , Survival Analysis , Virulence , Virus Internalization , Virus Replication
14.
J Infect Dis ; 207(3): 469-78, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23175762

ABSTRACT

Nipah virus (NiV) and Hendra virus (HeV) are closely related, recently emerged paramyxoviruses that are capable of causing considerable morbidity and mortality in several mammalian species, including humans. Henipavirus-specific vaccines are still commercially unavailable, and development of novel antiviral strategies to prevent lethal infections due to henipaviruses is highly desirable. Here we describe the development of adeno-associated virus (AAV) vaccines expressing the NiV G protein. Characterization of these vaccines in mice demonstrated that a single intramuscular AAV injection was sufficient to induce a potent and long-lasting antibody response. Translational studies in hamsters further demonstrated that all vaccinated animals were protected against lethal challenge with NiV. In addition, this vaccine induced a cross-protective immune response that was able to protect 50% of the animals against a challenge by HeV. This study presents a new efficient vaccination strategy against henipaviruses and opens novel perspectives on the use of AAV vectors as vaccines against emergent diseases.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Henipavirus Infections/immunology , Henipavirus Infections/prevention & control , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line, Tumor , Cricetinae , Disease Models, Animal , Henipavirus Infections/virology , Humans , Immunity, Humoral , Immunoglobulin G/immunology , Male , Mice , Vaccines, Synthetic/genetics , Viral Vaccines/genetics
15.
Emerg Microbes Infect ; 13(1): 2348508, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38661085

ABSTRACT

The Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne bunyavirus that causes high mortality in humans. This enveloped virus harbors two surface glycoproteins (GP), Gn and Gc, that are released by processing of a glycoprotein precursor complex whose maturation takes place in the ER and is completed through the secretion pathway. Here, we characterized the trafficking network exploited by CCHFV GPs during viral assembly, envelopment, and/or egress. We identified membrane trafficking motifs in the cytoplasmic domains (CD) of CCHFV GPs and addressed how they impact these late stages of the viral life cycle using infection and biochemical assays, and confocal microscopy in virus-producing cells. We found that several of the identified CD motifs modulate GP transport through the retrograde trafficking network, impacting envelopment and secretion of infectious particles. Finally, we identified PACS-2 as a crucial host factor contributing to CCHFV GPs trafficking required for assembly and release of viral particles.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo , Protein Transport , Virus Assembly , Humans , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Animals , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Protein Domains , Amino Acid Motifs , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Chlorocebus aethiops , HEK293 Cells , Vero Cells
16.
Nat Commun ; 15(1): 4542, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806525

ABSTRACT

The Crimean-Congo hemorrhagic fever virus (CCHFV) is an emerging pathogen of the Orthonairovirus genus that can cause severe and often lethal hemorrhagic diseases in humans. CCHFV has a broad tropism and can infect a variety of species and tissues. Here, by using gene silencing, blocking antibodies or soluble receptor fragments, we identify the low-density lipoprotein receptor (LDL-R) as a CCHFV entry factor. The LDL-R facilitates binding of CCHFV particles but does not allow entry of Hazara virus (HAZV), another member of the genus. In addition, we show that apolipoprotein E (apoE), an exchangeable protein that mediates LDL/LDL-R interaction, is incorporated on CCHFV particles, though not on HAZV particles, and enhances their specific infectivity by promoting an LDL-R dependent entry. Finally, we show that molecules that decrease LDL-R from the surface of target cells could inhibit CCHFV infection. Our study highlights that CCHFV takes advantage of a lipoprotein receptor and recruits its natural ligand to promote entry into cells.


Subject(s)
Apolipoproteins E , Hemorrhagic Fever Virus, Crimean-Congo , Receptors, LDL , Virus Internalization , Humans , Receptors, LDL/metabolism , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Hemorrhagic Fever Virus, Crimean-Congo/metabolism , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Animals , HEK293 Cells , Chlorocebus aethiops , Hemorrhagic Fever, Crimean/virology , Hemorrhagic Fever, Crimean/metabolism , Virion/metabolism , Vero Cells
17.
Antiviral Res ; 228: 105939, 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38909960

ABSTRACT

Viruses have developed sophisticated strategies to control metabolic activity of infected cells in order to supply replication machinery with energy and metabolites. Dengue virus (DENV), a mosquito-borne flavivirus responsible for dengue fever, is no exception. Previous reports have documented DENV interactions with metabolic pathways and shown in particular that glycolysis is increased in DENV-infected cells. However, underlying molecular mechanisms are still poorly characterized and dependence of DENV on this pathway has not been investigated in details yet. Here, we identified an interaction between the non-structural protein 3 (NS3) of DENV and glucokinase regulator protein (GCKR), a host protein that inhibits the liver-specific hexokinase GCK. NS3 expression was found to increase glucose consumption and lactate secretion in hepatic cell line expressing GCK. Interestingly, we observed that GCKR interaction with GCK decreases DENV replication, indicating the dependence of DENV to GCK activity and supporting the role of NS3 as an inhibitor of GCKR function. Accordingly, in the same cells, DENV replication both induces and depends on glycolysis. By targeting NAD(H) biosynthesis with the antimetabolite 6-Amino-Nicotinamide (6-AN), we decreased cellular glycolytic activity and inhibited DENV replication in hepatic cells. Infection of primary organotypic liver cultures (OLiC) from hamsters was also inhibited by 6-AN. Altogether, our results show that DENV has evolved strategies to control glycolysis in the liver, which could account for hepatic dysfunctions associated to infection. Besides, our findings suggest that lowering intracellular availability of NAD(H) could be a valuable therapeutic strategy to control glycolysis and inhibit DENV replication in the liver.

18.
Science ; 384(6703): eadm8693, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38935733

ABSTRACT

Measles virus (MeV) presents a public health threat that is escalating as vaccine coverage in the general population declines and as populations of immunocompromised individuals, who cannot be vaccinated, increase. There are no approved therapeutics for MeV. Neutralizing antibodies targeting viral fusion are one potential therapeutic approach but have not yet been structurally characterized or advanced to clinical use. We present cryo-electron microscopy (cryo-EM) structures of prefusion F alone [2.1-angstrom (Å) resolution], F complexed with a fusion-inhibitory peptide (2.3-Å resolution), F complexed with the neutralizing and protective monoclonal antibody (mAb) 77 (2.6-Å resolution), and an additional structure of postfusion F (2.7-Å resolution). In vitro assays and examination of additional EM classes show that mAb 77 binds prefusion F, arrests F in an intermediate state, and prevents transition to the postfusion conformation. These structures shed light on antibody-mediated neutralization that involves arrest of fusion proteins in an intermediate state.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , Antibodies, Viral , Cryoelectron Microscopy , Measles virus , Viral Fusion Proteins , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/chemistry , Measles virus/immunology , Measles virus/chemistry , Viral Fusion Proteins/immunology , Viral Fusion Proteins/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/chemistry , Humans , Protein Conformation
19.
J Virol ; 86(19): 10766-75, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22837207

ABSTRACT

Nipah virus (NiV) is a highly pathogenic, negative-strand RNA paramyxovirus that has recently emerged from flying foxes to cause serious human disease. We have analyzed the role of the nonstructural NiV C protein in viral immunopathogenesis using recombinant virus lacking the expression of NiV C (NiVΔC). While wild-type NiV was highly pathogenic in the hamster animal model, NiVΔC was strongly attenuated. Replication of NiVΔC was followed by the production of NiV-specific antibodies and associated with higher recruitment of inflammatory cells and less intensive histopathological lesions in different organs than in wild-type-NiV-infected animals. To analyze the molecular basis of NiVΔC attenuation, we studied early changes in gene expression in infected primary human endothelial cells, a major cellular target of NiV infection. The transcriptomic approach revealed the striking difference between wild-type and mutant NiV in the expression of genes involved in immunity, with the particularly interesting differential patterns of proinflammatory cytokines. Compared to wild-type virus, NiVΔC induced increased expression of interleukin 1 beta (IL-1ß), IL-8, CXCL2, CXCL3, CXCL6, CCL20, and beta interferon. Furthermore, the expression of NiV C in stably transfected cells decreased the production of the same panel of cytokines, revealing a role of the C protein in the regulation of cytokine balance. Together, these results suggest that NiV C regulates expression of proinflammatory cytokines, therefore providing a signal responsible for the coordination of leukocyte recruitment and the chemokine-induced immune response and controlling the lethal outcome of the infection.


Subject(s)
Phosphoproteins/genetics , Phosphoproteins/physiology , Viral Proteins/genetics , Viral Proteins/physiology , Animals , Cricetinae , Cytokines/metabolism , Endothelial Cells/cytology , Endothelial Cells/virology , Gene Expression Regulation, Viral , HEK293 Cells , Humans , Inflammation , Mesocricetus , Microcirculation , Nipah Virus/metabolism , Recombinant Proteins/chemistry , Time Factors , Umbilical Veins/cytology , Virulence
20.
Front Neurosci ; 17: 1108212, 2023.
Article in English | MEDLINE | ID: mdl-36937670

ABSTRACT

There is increasing evidence that viral infections are the source/origin of various types of encephalitis, encephalomyelitis, and other neurological and cognitive disorders. While the involvement of certain viruses, such as the Nipah virus and measles virus, is known, the mechanisms of neural invasion and the factors that trigger intense immune reactions are not fully understood. Based on recent publications, this review discusses the role of the immune response, interactions between viruses and glial cells, and cytokine mediators in the development of inflammatory diseases in the central nervous system. It also highlights the significant gaps in knowledge regarding these mechanisms.

SELECTION OF CITATIONS
SEARCH DETAIL