Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
1.
Clin Microbiol Infect ; 30(7): 930-936, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38552793

RÉSUMÉ

OBJECTIVES: The aim of this study was to assess the safety and immunogenicity of a dose-sparing fractional intradermal (ID) booster strategy with the mRNA-1273 COVID-19 vaccine. METHODS: COVID-19 naive adults aged 18-30 years were recruited from a previous study on primary vaccination regimens that compared 20 µg ID vaccinations with 100 µg intramuscular (IM) vaccinations with mRNA-1273 as the primary vaccination series. Participants previously immunized with ID regimens were randomly assigned (1:1) to receive a fractional ID booster dose (20 µg) or the standard-of-care intramuscular (IM) booster dose (50 µg) of the mRNA-1273 vaccine, 6 months after completing their primary series (ID-ID and ID-IM group, respectively). Participants that had received a full dose IM regimen as the primary series, received the IM standard-of-care booster dose (IM-IM group). In addition, COVID-19 naive individuals aged 18-40 years who had received an IM mRNA vaccine as the primary series were recruited from the general population to receive a fractional ID booster dose (IM-ID group). Immunogenicity was assessed using IgG anti-spike antibody responses and neutralizing capacity against SARS-CoV-2. Cellular immune responses were measured in a sub-group. Safety and tolerability were monitored. RESULTS: In January 2022, 129 participants were included in the study. Fractional ID boosting was safe and well tolerated, with fewer systemic adverse events compared with IM boosting. At day 28 post-booster, anti-spike S1 IgG geometric mean concentrations were 9106 (95% CI, 7150-11 597) binding antibody units (BAU)/mL in the IM-IM group and 4357 (3003-6322) BAU/mL; 6629 (4913-8946) BAU/mL; and 5264 (4032-6873) BAU/mL in the ID-IM, ID-ID, and IM-ID groups, respectively. DISCUSSION: Intradermal boosting provides robust immune responses and is a viable dose-sparing strategy for mRNA COVID-19 vaccines. The favourable side-effect profile supports its potential to reduce vaccine hesitancy. Fractional dosing strategies should be considered early in the clinical development of future mRNA vaccines to enhance vaccine availability and pandemic preparedness.


Sujet(s)
Vaccin ARNm-1273 contre la COVID-19 , Anticorps neutralisants , Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , Rappel de vaccin , Immunogénicité des vaccins , SARS-CoV-2 , Humains , Adulte , Rappel de vaccin/méthodes , Injections intradermiques , Mâle , Femelle , COVID-19/prévention et contrôle , COVID-19/immunologie , Jeune adulte , Anticorps antiviraux/sang , SARS-CoV-2/immunologie , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Vaccins contre la COVID-19/effets indésirables , Anticorps neutralisants/sang , Adolescent , Injections musculaires , Vaccination/méthodes
2.
PLoS Pathog ; 20(3): e1012100, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38527094

RÉSUMÉ

The coronavirus papain-like protease (PLpro) is crucial for viral replicase polyprotein processing. Additionally, PLpro can subvert host defense mechanisms by its deubiquitinating (DUB) and deISGylating activities. To elucidate the role of these activities during SARS-CoV-2 infection, we introduced mutations that disrupt binding of PLpro to ubiquitin or ISG15. We identified several mutations that strongly reduced DUB activity of PLpro, without affecting viral polyprotein processing. In contrast, mutations that abrogated deISGylating activity also hampered viral polyprotein processing and when introduced into the virus these mutants were not viable. SARS-CoV-2 mutants exhibiting reduced DUB activity elicited a stronger interferon response in human lung cells. In a mouse model of severe disease, disruption of PLpro DUB activity did not affect lethality, virus replication, or innate immune responses in the lungs. This suggests that the DUB activity of SARS-CoV-2 PLpro is dispensable for virus replication and does not affect innate immune responses in vivo. Interestingly, the DUB mutant of SARS-CoV replicated to slightly lower titers in mice and elicited a diminished immune response early in infection, although lethality was unaffected. We previously showed that a MERS-CoV mutant deficient in DUB and deISGylating activity was strongly attenuated in mice. Here, we demonstrate that the role of PLpro DUB activity during infection can vary considerably between highly pathogenic coronaviruses. Therefore, careful considerations should be taken when developing pan-coronavirus antiviral strategies targeting PLpro.


Sujet(s)
COVID-19 , Protéases de type papaïne des coronavirus , Humains , Animaux , Souris , Protéases de type papaïne des coronavirus/génétique , SARS-CoV-2/métabolisme , Immunité innée , Papaïne/génétique , Papaïne/métabolisme , Peptide hydrolases/métabolisme , Réplication virale , Polyprotéines
3.
Viruses ; 16(2)2024 02 05.
Article de Anglais | MEDLINE | ID: mdl-38400032

RÉSUMÉ

Mitochondrial antiviral signaling protein (MAVS) is a crucial signaling adaptor in the sensing of positive-sense RNA viruses and the subsequent induction of the innate immune response. Coronaviruses have evolved multiple mechanisms to evade this response, amongst others, through their main protease (Mpro), which is responsible for the proteolytic cleavage of the largest part of the viral replicase polyproteins pp1a and pp1ab. Additionally, it can cleave cellular substrates, such as innate immune signaling factors, to dampen the immune response. Here, we show that MAVS is cleaved in cells infected with Middle East respiratory syndrome coronavirus (MERS-CoV), but not in cells infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This cleavage was independent of cellular negative feedback mechanisms that regulate MAVS activation. Furthermore, MERS-CoV Mpro expression induced MAVS cleavage upon overexpression and suppressed the activation of the interferon-ß (IFN-ß) and nuclear factor-κB (NF-κB) response. We conclude that we have uncovered a novel mechanism by which MERS-CoV downregulates the innate immune response, which is not observed among other highly pathogenic coronaviruses.


Sujet(s)
Coronavirus du syndrome respiratoire du Moyen-Orient , Immunité innée , Interféron bêta/métabolisme , Peptide hydrolases , Antiviraux
4.
J Virol ; 90(22): 10209-10219, 2016 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-27581985

RÉSUMÉ

Due to antigenic drift of influenza viruses, seasonal influenza vaccines need to be updated annually. These vaccines are based on predictions of strains likely to circulate in the next season. However, vaccine efficacy is greatly reduced in the case of a mismatch between circulating and vaccine strains. Furthermore, novel antigenically distinct influenza viruses are introduced into the human population from animal reservoirs occasionally and may cause pandemic outbreaks. To dampen the impact of seasonal and pandemic influenza, vaccines that induce broadly protective and long-lasting immunity are preferred. Because influenza virus-specific CD8+ T cells are directed mainly against relatively conserved internal proteins, like nucleoprotein (NP), they are highly cross-reactive and afford protection against infection with antigenically distinct influenza virus strains, so-called heterosubtypic immunity. Here, we used modified vaccinia virus Ankara (MVA) as a vaccine vector for the induction of influenza virus NP-specific CD8+ T cells. To optimize the induction of CD8+ T cell responses, we made several modifications to NP, aiming at retaining the protein in the cytosol or targeting it to the proteasome. We hypothesized that these strategies would increase antigen processing and presentation and thus improve the induction of CD8+ T cell responses. We showed that NP with increased degradation rates improved CD8+ T cell activation in vitro if the amount of antigen was limited or if CD8+ T cells were of low functional avidity. However, after immunization of C57BL/6 mice, no differences were detected between modified NP and wild-type NP (NPwt), since NPwt already induced optimal CD8+ T cell responses. IMPORTANCE: Due to the continuous antigenic drift of seasonal influenza viruses and the threat of a novel pandemic, there is a great need for the development of novel influenza vaccines that offer broadly protective immunity against multiple subtypes. CD8+ T cells can provide immunity against multiple subtypes of influenza viruses by the recognition of relatively conserved internal antigens. In this study, we aimed at optimizing the CD8+ T cell response to influenza A virus by making modifications to influenza A virus nucleoprotein (NP) expressed from the modified vaccinia virus Ankara (MVA) vaccine vector. These modifications resulted in increased antigen degradation, thereby producing elevated levels of peptides that can be presented on major histocompatibility complex (MHC) class I molecules to CD8+ T cells. Although we were unable to increase the NP-specific immune response in the mouse strain used, this approach may have benefits for vaccine development using less-immunogenic proteins.


Sujet(s)
Lymphocytes T CD8+/immunologie , Virus de la grippe A/immunologie , Virus de la grippe A/métabolisme , Activation des lymphocytes/immunologie , Protéines de liaison à l'ARN/métabolisme , Protéines du core viral/métabolisme , Animaux , Anticorps antiviraux/métabolisme , Antigènes viraux/immunologie , Lignée cellulaire , Lignée cellulaire tumorale , Poulets , Réactions croisées/immunologie , Chiens , Femelle , Cellules HeLa , Humains , Vaccins antigrippaux/immunologie , Cellules rénales canines Madin-Darby , Souris , Souris de lignée C57BL , Protéines nucléocapside , Infections à Orthomyxoviridae/virologie , Protéolyse , Protéines de liaison à l'ARN/immunologie , Vaccination/méthodes , Virus de la vaccine/immunologie , Protéines du core viral/immunologie
6.
J Infect Dis ; 211(5): 791-800, 2015 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-25246535

RÉSUMÉ

Since the first reports in early 2013, >440 human cases of infection with avian influenza A(H7N9) have been reported including 122 fatalities. After the isolation of the first A(H7N9) viruses, the nucleotide sequences became publically available. Based on the coding sequence of the influenza virus A/Shanghai/2/2013 hemagglutinin gene, a codon-optimized gene was synthesized and cloned into a recombinant modified vaccinia virus Ankara (MVA). This MVA-H7-Sh2 viral vector was used to immunize ferrets and proved to be immunogenic, even after a single immunization. Subsequently, ferrets were challenged with influenza virus A/Anhui/1/2013 via the intratracheal route. Unprotected animals that were mock vaccinated or received empty vector developed interstitial pneumonia characterized by a marked alveolitis, accompanied by loss of appetite, weight loss, and heavy breathing. In contrast, animals vaccinated with MVA-H7-Sh2 were protected from severe disease.


Sujet(s)
Vecteurs de médicaments , Vecteurs génétiques , Glycoprotéine hémagglutinine du virus influenza/immunologie , Sous-type H7N9 du virus de la grippe A/immunologie , Vaccins antigrippaux/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Virus de la vaccine/génétique , Animaux , Modèles animaux de maladie humaine , Femelle , Furets , Glycoprotéine hémagglutinine du virus influenza/génétique , Sous-type H7N9 du virus de la grippe A/génétique , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/génétique , Pneumopathies interstitielles/anatomopathologie , Pneumopathies interstitielles/prévention et contrôle , Infections à Orthomyxoviridae/anatomopathologie , Résultat thérapeutique , Vaccination/méthodes
7.
Lancet Infect Dis ; 14(12): 1196-207, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25455987

RÉSUMÉ

BACKGROUND: Modified vaccinia virus Ankara (MVA) is a promising viral vector platform for the development of an H5N1 influenza vaccine. Preclinical assessment of MVA-based H5N1 vaccines showed their immunogenicity and safety in different animal models. We aimed to assess the safety and immunogenicity of the MVA-haemagglutinin-based H5N1 vaccine MVA-H5-sfMR in healthy individuals. METHODS: In a single-centre, double-blind phase 1/2a study, young volunteers (aged 18-28 years) were randomly assigned with a computer-generated list in equal numbers to one of eight groups and were given one injection or two injections intramuscularly at an interval of 4 weeks of a standard dose (10(8) plaque forming units [pfu]) or a ten times lower dose (10(7) pfu) of the MVA-H5-sfMR (vector encoding the haemagglutinin gene of influenza A/Vietnam/1194/2004 virus [H5N1 subtype]) or MVA-F6-sfMR (empty vector) vaccine. Volunteers and physicians who examined and administered the vaccine were masked to vaccine assignment. Individuals who received the MVA-H5-sfMR vaccine were eligible for a booster immunisation 1 year after the first immunisation. Primary endpoint was safety. Secondary outcome was immunogenicity. The trial is registered with the Dutch Trial Register, number NTR3401. FINDINGS: 79 of 80 individuals who were enrolled completed the study. No serious adverse events were identified. 11 individuals reported severe headache and lightheadedness, erythema nodosum, respiratory illness (accompanied by influenza-like symptoms), sore throat, or injection-site reaction. Most of the volunteers had one or more local (itch, pain, redness, and swelling) and systemic reactions (rise in body temperature, headache, myalgia, arthralgia, chills, malaise, and fatigue) after the first, second, and booster immunisations. Individuals who received the 10(7) dose had fewer systemic reactions. The MVA-H5-sfMR vaccine at 10(8) pfu induced significantly higher antibody responses after one and two immunisations than did 10(7) pfu when assessed with haemagglutination inhibition geometric mean titre at 8 weeks against H5N1 A/Vietnam/1194/2004 (30·2 [SD 3·8] vs 9·2 [2·3] and 108·1 [2·4] vs 15·8 [3·2]). 27 of 39 eligible individuals were enrolled in the booster immunisation study. A single shot of MVA-H5-sfMR 10(8) pfu prime immunisation resulted in higher antibody responses after the booster immunisation than did two shots of MVA-H5-sfMR at the ten times lower dose. INTERPRETATION: The MVA-based H5N1 vaccine was well tolerated and immunogenic and therefore the vaccine candidates arising from the MVA platform hold great promise for rapid development in response to a future influenza pandemic threat. However, the immunogenicity of this vaccine needs to be compared with conventional H5N1 inactivated non-adjuvanted vaccine candidates in head-to-head clinical trials. FUNDING: European Research Council.


Sujet(s)
Vecteurs de médicaments , Vecteurs génétiques , Glycoprotéine hémagglutinine du virus influenza/immunologie , Sous-type H5N1 du virus de la grippe A/immunologie , Vaccins antigrippaux/effets indésirables , Vaccins antigrippaux/immunologie , Virus de la vaccine/génétique , Adolescent , Adulte , Animaux , Anticorps antiviraux/sang , Méthode en double aveugle , Effets secondaires indésirables des médicaments/épidémiologie , Effets secondaires indésirables des médicaments/anatomopathologie , Femelle , Volontaires sains , Tests d'inhibition de l'hémagglutination , Glycoprotéine hémagglutinine du virus influenza/génétique , Humains , Sous-type H5N1 du virus de la grippe A/génétique , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/génétique , Injections musculaires , Mâle , Vaccins synthétiques/administration et posologie , Vaccins synthétiques/effets indésirables , Vaccins synthétiques/génétique , Vaccins synthétiques/immunologie , Jeune adulte
8.
Neurobiol Aging ; 33(9): 2210-4, 2012 Sep.
Article de Anglais | MEDLINE | ID: mdl-21925769

RÉSUMÉ

Alzheimer's disease (AD) is the most common form of dementia, increasing in prevalence with age. Most patients who develop AD have an unknown cause, but characteristic neuropathological features include the deposition of extracellular amyloid beta and of intraneuronal hyperphosphorylated tau protein. Researchers have previously implicated mitochondrial dysfunction in AD. We previously showed an increase in neurons displaying a mitochondrial biochemical defect-cytochrome-c oxidase (COX) deficiency-in the hippocampus in patients with sporadic AD compared with age-matched controls. COX deficiency is well described as a marker of mitochondrial (mt) DNA dysfunction. This present study analyzed the mtDNA in single neurons from both COX normal and COX-deficient cells. Analysis of the mtDNA revealed that COX deficiency is caused by high levels of mtDNA deletions which accumulate with age. Future research is needed to clarify the role mtDNA deletions have in normal aging and investigate the relationship between mtDNA deletions and the pathogenesis of sporadic AD.


Sujet(s)
Maladie d'Alzheimer/complications , Maladie d'Alzheimer/génétique , Déficit en cytochrome-c oxydase , Neurones/enzymologie , Délétion de séquence/génétique , Sujet âgé , Études cas-témoins , Déficit en cytochrome-c oxydase/étiologie , Déficit en cytochrome-c oxydase/génétique , Déficit en cytochrome-c oxydase/anatomopathologie , Complexe IV de la chaîne respiratoire/métabolisme , Femelle , Humains , Mâle , Mitochondries/enzymologie , Mitochondries/anatomopathologie , NADH dehydrogenase/génétique , Modifications postmortem , ARN messager/métabolisme
9.
Clin Chem Lab Med ; 45(11): 1555-8, 2007.
Article de Anglais | MEDLINE | ID: mdl-17924844

RÉSUMÉ

BACKGROUND: Carbohydrate-deficient transferrin (CDT) is a biochemical marker used for identifying chronic alcohol intake. We developed and validated an ARCHITECT c8000 (Abbott) instrument application for the Axis-Shield %CDT immunoassay. METHODS: Standard CLSI (Clinical and Laboratory Standards Institute) evaluation protocols were performed. RESULTS: The Axis-Shield %CDT ARCHITECT method was standardized against the Axis-Shield %CDT microtiter test by linear regression analysis (n=50 mean of duplicate, R=0.996). Method comparison by Deming regression revealed a slope of 1.01 and an intercept of -0.03 with Axis-Shield %CDT microtiter test (n=50 in duplicate, R=0.995), and a slope of 0.82 and an intercept of 1.09 with HPLC method (n=47 in duplicate, R=0.990) as the candidate IFCC (International Federation of Clinical Chemistry and Laboratory Medicine) reference method. The predicted medical decision points (MDPs) are both 2.6% and equal the MDP that is generally used for the Axis-Shield %CDT tests. Precision derived from pooled patient sera (low level) and commercially available control material (high level) was excellent. Total variation was 3.2% and 1.8%, respectively. CONCLUSIONS: The Axis-Shield %CDT ARCHITECT method, as one of the first Axis-Shield applications on a large-scale analyzer, is a reliable test for routine %CDT analysis providing precise and well-standardized %CDT results.


Sujet(s)
Alcoolisme/diagnostic , Transferrine/analogues et dérivés , Alcoolisme/sang , Calibrage , Chromatographie en phase liquide à haute performance , Humains , Transferrine/analyse
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...