Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 304
Filtrar
1.
Nature ; 626(7998): 392-400, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38086420

RESUMO

An ideal vaccine both attenuates virus growth and disease in infected individuals and reduces the spread of infections in the population, thereby generating herd immunity. Although this strategy has proved successful by generating humoral immunity to measles, yellow fever and polio, many respiratory viruses evolve to evade pre-existing antibodies1. One approach for improving the breadth of antiviral immunity against escape variants is through the generation of memory T cells in the respiratory tract, which are positioned to respond rapidly to respiratory virus infections2-6. However, it is unknown whether memory T cells alone can effectively surveil the respiratory tract to the extent that they eliminate or greatly reduce viral transmission following exposure of an individual to infection. Here we use a mouse model of natural parainfluenza virus transmission to quantify the extent to which memory CD8+ T cells resident in the respiratory tract can provide herd immunity by reducing both the susceptibility of acquiring infection and the extent of transmission, even in the absence of virus-specific antibodies. We demonstrate that protection by resident memory CD8+ T cells requires the antiviral cytokine interferon-γ (IFNγ) and leads to altered transcriptional programming of epithelial cells within the respiratory tract. These results suggest that tissue-resident CD8+ T cells in the respiratory tract can have important roles in protecting the host against viral disease and limiting viral spread throughout the population.


Assuntos
Linfócitos T CD8-Positivos , Memória Imunológica , Células T de Memória , Infecções por Paramyxoviridae , Sistema Respiratório , Animais , Camundongos , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Imunidade Coletiva/imunologia , Memória Imunológica/imunologia , Interferon gama/imunologia , Células T de Memória/imunologia , Paramyxoviridae/imunologia , Paramyxoviridae/fisiologia , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Infecções por Paramyxoviridae/transmissão , Infecções por Paramyxoviridae/virologia , Sistema Respiratório/citologia , Sistema Respiratório/imunologia , Sistema Respiratório/virologia , Transcrição Gênica , Humanos
2.
Nat Commun ; 14(1): 798, 2023 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-36781872

RESUMO

Respiratory syncytial virus (RSV), human metapneumovirus (HMPV), and human parainfluenza virus types one (HPIV1) and three (HPIV3) can cause severe disease and death in immunocompromised patients, the elderly, and those with underlying lung disease. A protective monoclonal antibody exists for RSV, but clinical use is limited to high-risk infant populations. Hence, therapeutic options for these viruses in vulnerable patient populations are currently limited. Here, we present the discovery, in vitro characterization, and in vivo efficacy testing of two cross-neutralizing monoclonal antibodies, one targeting both HPIV3 and HPIV1 and the other targeting both RSV and HMPV. The 3 × 1 antibody is capable of targeting multiple parainfluenza viruses; the MxR antibody shares features with other previously reported monoclonal antibodies that are capable of neutralizing both RSV and HMPV. We obtained structures using cryo-electron microscopy of these antibodies in complex with their antigens at 3.62 Å resolution for 3 × 1 bound to HPIV3 and at 2.24 Å for MxR bound to RSV, providing a structural basis for in vitro binding and neutralization. Together, a cocktail of 3 × 1 and MxR could have clinical utility in providing broad protection against four of the respiratory viruses that cause significant morbidity and mortality in at-risk individuals.


Assuntos
Metapneumovirus , Infecções por Paramyxoviridae , Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Humanos , Anticorpos Monoclonais , Anticorpos Neutralizantes , Anticorpos Antivirais , Microscopia Crioeletrônica , Infecções por Paramyxoviridae/prevenção & controle , Proteínas Virais de Fusão , Proteção Cruzada
3.
Viruses ; 14(10)2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-36298690

RESUMO

Non-pharmaceutical interventions (NPIs) to reduce SARS-CoV-2 transmission disrupted respiratory virus seasonality. We examined the unusual return of human metapneumovirus (hMPV) in Western Australia following a period of absence in 2020. We analysed hMPV laboratory testing data from 1 January 2017 to 31 December 2021. Whole-genome sequencing of selected hMPV-positive samples was performed using a tiled-amplicon approach. Following an absence in spring 2020, an unusual hMPV surge was observed during the wet summer season in the tropical Northern region in late 2020. Following a six-month delay, an intense winter season occurred in the subtropical/temperate Southern and Metropolitan regions. Compared to 2017-2019, hMPV incidence in 2021 increased by 3-fold, with a greater than 4-fold increase in children aged 1-4 years. There was a collapse in hMPV diversity in 2020, with the emergence of a single subtype. NPIs contributed to an absent 2020 season and a clonal hMPV resurgence. The summer surge and delayed winter season suggest that prevailing temperature and humidity are keys determinant of hMPV transmission. The increased incidence in 2021 was linked to an expanded cohort of hMPV-naïve 1-4-year-old children and waning population immunity. Further intense and unusual respiratory virus seasons are expected as COVID-19 associated NPIs are removed.


Assuntos
COVID-19 , Metapneumovirus , Infecções por Paramyxoviridae , Infecções Respiratórias , Humanos , Lactente , Pré-Escolar , Metapneumovirus/genética , Infecções por Paramyxoviridae/epidemiologia , Infecções por Paramyxoviridae/prevenção & controle , SARS-CoV-2/genética , Austrália Ocidental/epidemiologia , COVID-19/epidemiologia , COVID-19/prevenção & controle , Estações do Ano
4.
Proc Natl Acad Sci U S A ; 119(25): e2203326119, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35696580

RESUMO

Human metapneumovirus (hMPV) is a leading cause of morbidity and hospitalization among children worldwide, however, no vaccines or therapeutics are currently available for hMPV disease prevention and treatment. The hMPV fusion (F) protein is the sole target of neutralizing antibodies. To map the immunodominant epitopes on the hMPV F protein, we isolated a panel of human monoclonal antibodies (mAbs), and the mAbs were assessed for binding avidity, neutralization potency, and epitope specificity. We found the majority of the mAbs target diverse epitopes on the hMPV F protein, and we discovered multiple mAb binding approaches for antigenic site III. The most potent mAb, MPV467, which had picomolar potency, was examined in prophylactic and therapeutic mouse challenge studies, and MPV467 limited virus replication in mouse lungs when administered 24 h before or 72 h after viral infection. We determined the structure of MPV467 in complex with the hMPV F protein using cryo-electron microscopy to a resolution of 3.3 Å, which revealed a complex novel prefusion-specific epitope overlapping antigenic sites II and V on a single protomer. Overall, our data reveal insights into the immunodominant antigenic epitopes on the hMPV F protein, identify a mAb therapy for hMPV F disease prevention and treatment, and provide the discovery of a prefusion-specific epitope on the hMPV F protein.


Assuntos
Anticorpos Monoclonais , Anticorpos Neutralizantes , Anticorpos Antivirais , Antígenos Virais , Metapneumovirus , Infecções por Paramyxoviridae , Proteínas Virais de Fusão , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/isolamento & purificação , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/química , Anticorpos Antivirais/isolamento & purificação , Anticorpos Antivirais/uso terapêutico , Antígenos Virais/química , Antígenos Virais/imunologia , Microscopia Crioeletrônica , Epitopos/imunologia , Humanos , Metapneumovirus/imunologia , Camundongos , Infecções por Paramyxoviridae/prevenção & controle , Prevenção Primária , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/imunologia
5.
Avian Pathol ; 51(2): 181-196, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35099352

RESUMO

Avian metapneumovirus (aMPV) causes respiratory disease and drops in egg production in chickens, and is routinely controlled by vaccination. However, the host's immune response to virulent challenge in vaccinated or unvaccinated broiler chickens is poorly characterized. We show that subtype B vaccination offers heterologous (subtype A challenge) and homologous (subtype B challenge) protection. Subtype B challenge caused significantly greater humoral antibody titres in vaccinated and unvaccinated chickens. In turbinate and lung tissues of unvaccinated-challenged chickens, IgA and IgY mRNA transcription was significantly up-regulated after subtype B challenge compared to subtype A. Cellular immunity (CD8-α and CD8-ß) gene transcripts were significantly up-regulated during early and later stages of infection from subtype B or subtype A, respectively. Immune gene transcriptional responses (IL-1ß, IL-6 and IL-18) were significantly up-regulated after challenge. Gene transcription results showed that mRNA expression levels of CD8-α, CD8-ß, TLR3 and IL-6, particularly in turbinate and trachea tissues, are useful parameters to include in future aMPV vaccination-challenge studies.


Assuntos
Metapneumovirus , Infecções por Paramyxoviridae , Doenças das Aves Domésticas , Animais , Anticorpos Antivirais , Galinhas , Imunidade Celular , Metapneumovirus/genética , Infecções por Paramyxoviridae/prevenção & controle , Infecções por Paramyxoviridae/veterinária , Vacinação/veterinária
6.
Sci Rep ; 11(1): 20769, 2021 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-34675220

RESUMO

Human metapneumovirus (HMPV) is a leading cause of acute lower respiratory tract illness in children and adults. Repeated infections are common and can be severe in young, elderly, and immunocompromised persons due to short-lived protective humoral immunity. In turn, few protective T cell epitopes have been identified in humans. Thus, we infected transgenic mice expressing the common human HLA MHC-I allele B*07:02 (HLA-B7) with HMPV and screened a robust library of overlapping and computationally predicted HLA-B7 binding peptides. Six HLA-B7-restricted CD8+ T cell epitopes were identified using ELISPOT screening in the F, M, and N proteins, with M195-203 (M195) eliciting the strongest responses. MHC-tetramer flow cytometric staining confirmed HLA-B7 epitope-specific CD8+ T cells migrated to lungs and spleen of HMPV-immune mice. Immunization with pooled HLA-B7-restricted peptides reduced viral titer and protected mice from virulent infection. Finally, we confirmed that CD8+ T cells from HLA-B7 positive humans also recognize the identified epitopes. These results enable identification of HMPV-specific CD8+ T cells in humans and help to inform future HMPV vaccine design.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Antígeno HLA-B7/imunologia , Metapneumovirus/imunologia , Infecções por Paramyxoviridae/imunologia , Animais , Células Cultivadas , Epitopos de Linfócito T/uso terapêutico , Humanos , Interferon gama/imunologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infecções por Paramyxoviridae/prevenção & controle , Peptídeos/imunologia , Peptídeos/uso terapêutico , Vacinas Virais/imunologia , Vacinas Virais/uso terapêutico
7.
Poult Sci ; 100(3): 100528, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33518307

RESUMO

This article describes a series of animal studies for the development of an avian metapneumovirus (aMPV) live vaccine. Although aMPV causes continual economic loss in the poultry industry, there are no live aMPV vaccines available in Korea. Furthermore, information is limited with respect to standard field practices for vaccinations at an early age. Here, the development of an aMPV live vaccine was attempted, and its efficacy was investigated with respect to the vaccination route and age to develop a method for controlling aMPV. Before vaccine development, an animal challenge model was established using the aMPV field isolate to identify the most effective time and site for collecting samples for evaluation. After attenuation of the virulent aMPV in Vero cells, a safety and efficacy test was conducted for the vaccine candidate. As a novel aMPV live vaccine candidate, aMPV K655/07HP displayed sufficient safety in day-old chicks with 10 vaccine doses. The efficacy test using 1-week-old chicks showed weaker humoral immune response than that in 4-week-old chicks. However, the candidate vaccine provided complete protection against infection caused by the challenge virus for all ages of vaccinated chicks. In conclusion, an effective aMPV challenge model was established for studying aMPV in chickens, which offered important, insightful information. The safety and efficacy study suggested that the new aMPV candidate vaccine could be used to effectively reduce the economic losses incurred because of aMPV infection.


Assuntos
Metapneumovirus , Infecções por Paramyxoviridae , Doenças das Aves Domésticas , Vacinas Virais , Fatores Etários , Animais , Anticorpos Antivirais/sangue , Galinhas/imunologia , Chlorocebus aethiops , Metapneumovirus/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Infecções por Paramyxoviridae/veterinária , Doenças das Aves Domésticas/prevenção & controle , República da Coreia , Vacinação/normas , Vacinação/veterinária , Vacinas Atenuadas/imunologia , Células Vero , Vacinas Virais/imunologia , Vacinas Virais/normas
8.
Vaccine ; 38(9): 2122-2127, 2020 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-32007293

RESUMO

Human metapneumovirus (hMPV) is an important respiratory pathogen especially in young children and elderly subjects. Our objective was to assess the immunogenicity and protection conferred by predominant pre- and post-fusion (F) hMPV-F constructs in Balb/C mice. Immunizations without adjuvant were not immunogenic whereas alum-adjuvanted hMPV-F proteins, regardless of their conformations, generated comparable neutralizing antibody titers with undetectable pulmonary viral titers following viral challenge. In conclusion, we found no apparent advantage for mixtures of predominant pre-fusion F proteins over post-fusion conformations for hMPV vaccination in opposite to recent data obtained with the human respiratory syncytial virus.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Metapneumovirus , Infecções por Paramyxoviridae , Proteínas Virais de Fusão/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Metapneumovirus/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Paramyxoviridae/prevenção & controle , Vacinas de Subunidades Antigênicas/imunologia , Proteínas Virais de Fusão/administração & dosagem
9.
Expert Rev Vaccines ; 19(1): 11-24, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31903811

RESUMO

Introduction: Influenza virus, human respiratory syncytial virus (RSV), and human metapneumovirus (HMPV) are important human respiratory pathogens. Recombinant virus-like particle (VLP) vaccines are suggested to be potential promising platforms to protect against these respiratory viruses. This review updates important progress in the development of VLP vaccines against respiratory viruses.Areas Covered: This review summarizes progress in developing VLP and nanoparticle-based vaccines against influenza virus, RSV, and HMPV. The PubMed was mainly used to search for important research articles published since 2010 although earlier key articles were also referenced. The research area covered includes VLP and nanoparticle platform vaccines against seasonal, pandemic, and avian influenza viruses as well as RSV and HMPV respiratory viruses. The production methods, immunogenic properties, and vaccine efficacy of respiratory VLP vaccines in preclinical animal models and clinical studies were reviewed in this article.Expert opinion: Previous and current preclinical and clinical studies suggest that recombinant VLP and nanoparticle vaccines are expected to be developed as promising alternative platforms against respiratory viruses in future. Therefore, continued research efforts are warranted.


Assuntos
Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas Virais/administração & dosagem , Animais , Humanos , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Nanopartículas , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia
10.
Front Immunol ; 11: 611946, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33584692

RESUMO

Vaccines represent an important strategy to protect humans against a wide variety of pathogens and have even led to eradicating some diseases. Although every vaccine is developed to induce specific protection for a particular pathogen, some vaccine formulations can also promote trained immunity, which is a non-specific memory-like feature developed by the innate immune system. It is thought that trained immunity can protect against a wide variety of pathogens other than those contained in the vaccine formulation. The non-specific memory of the trained immunity-based vaccines (TIbV) seems beneficial for the immunized individual, as it may represent a powerful strategy that contributes to the control of pathogen outbreaks, reducing morbidity and mortality. A wide variety of respiratory viruses, including respiratory syncytial virus (hRSV) and metapneumovirus (hMPV), cause serious illness in children under 5 years old and the elderly. To address this public health problem, we have developed recombinant BCG vaccines that have shown to be safe and immunogenic against hRSV or hMPV. Besides the induction of specific adaptive immunity against the viral antigens, these vaccines could generate trained immunity against other respiratory pathogens. Here, we discuss some of the features of trained immunity induced by BCG and put forward the notion that recombinant BCGs expressing hRSV or hMPV antigens have the capacity to simultaneously induce specific adaptive immunity and non-specific trained immunity. These recombinant BCG vaccines could be considered as TIbV capable of inducing simultaneously the development of specific protection against hRSV or hMPV, as well as non-specific trained-immunity-based protection against other pathogenic viruses.


Assuntos
Vacina BCG/administração & dosagem , Imunogenicidade da Vacina , Metapneumovirus/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Animais , Vacina BCG/imunologia , Humanos , Imunidade Inata , Memória Imunológica , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/virologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vacinas contra Vírus Sincicial Respiratório/imunologia , Resultado do Tratamento , Vacinação , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
11.
J Health Commun ; 25(1): 33-42, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31868126

RESUMO

Answering the call by some health communication researchers to give greater attention to message strategies at the level of word choices and sentence structures, this study examined how the linguistic marking of argumentative orientation and linguistic agency assignment affects young adults' reactions to an informational message about a sexually transmitted infection presented as a new emerging health threat. Participants were randomly assigned to read one of the four versions of a fact sheet defined by a 2 × 2 (agency assignment x marking of argumentation orientation) factorial design and thereafter completed a questionnaire. Results indicated that the assignment of agency to the virus (vs. human) increased the perceived severity of the health threat, perceived susceptibility to it, persuasiveness of the message, and safer sex intentions. The same outcomes occurred when the message was phrased with a high marking of the argumentative orientation rather than a low marking. These findings suggest that a better understanding of language variable effects can boost the efficacy of promotional health messages.


Assuntos
Doenças Transmissíveis Emergentes/prevenção & controle , Comunicação em Saúde/métodos , Linguística , Infecções por Paramyxoviridae/prevenção & controle , Comunicação Persuasiva , Infecções Sexualmente Transmissíveis/prevenção & controle , Adolescente , Feminino , França/epidemiologia , Conhecimentos, Atitudes e Prática em Saúde , Humanos , Masculino , Infecções por Paramyxoviridae/epidemiologia , Infecções Sexualmente Transmissíveis/epidemiologia , Estudantes/psicologia , Estudantes/estatística & dados numéricos , Universidades , Adulto Jovem
12.
Poult Sci ; 98(11): 5374-5384, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31264704

RESUMO

Respiratory diseases like infectious bronchitis virus (IBV) and avian metapneumovirus (aMPV) have been held accountable for major losses for poultry production. Nevertheless, scarce information was present dealing with the prevalence and molecular epidemiology of these infections in Greece and the efficacy of currently applied control strategies. To fill this gap, a specific epidemiological study was designed. A total of 106 broiler and layer farms, including 10 backyard and 96 commercial flocks, were sampled between March 2016 and May 2017, and the obtained tracheal swabs were tested for IBV and aMPV using RT-PCR based techniques followed by sequencing. For each farm, data regarding production type, flock features, clinical signs, and vaccination program were also recorded. Different associations between vaccination protocol, production type, animal category, birds density, age, presence of clinical signs, and IBV and/or aMPV infection were tested. Both IBV and aMPV field strain prevalence were proven high, approximately 20 and 30%, respectively, being the GI-19 lineage (14 out of 19; 73.6%) and B subtype (30 out of 30; 100%), the most commonly detected IBV and aMPV genetic types. Infection with IBV field strains was significantly associated with clinical sign presence (odds ratio = 8.55 [95CI = 2.17-42.90]). Remarkably, only the vaccination protocol involving a double vaccination at 1 D of age was proven protective against IBV-induced symptomatology, with the odds of developing disease being 4.14 [95CI = 1.34-14.51] times lower. No association was demonstrated between aMPV infection and clinical outbreaks or between aMPV and IBV detection, suggesting the marginal role of the former pathogen in poultry farming. Globally, the present study provides the first detailed investigation of the epidemiological scenario of 2 viruses traditionally considered of pivotal relevance in poultry farming and demonstrates that remarkable benefits could be obtained with just minor adjustments in vaccination protocols.


Assuntos
Galinhas , Infecções por Coronavirus/veterinária , Vírus da Bronquite Infecciosa/fisiologia , Metapneumovirus/fisiologia , Infecções por Paramyxoviridae/veterinária , Doenças das Aves Domésticas/epidemiologia , Animais , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/virologia , Grécia/epidemiologia , Vírus da Bronquite Infecciosa/genética , Metapneumovirus/genética , Epidemiologia Molecular , Infecções por Paramyxoviridae/epidemiologia , Infecções por Paramyxoviridae/prevenção & controle , Infecções por Paramyxoviridae/virologia , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Prevalência , Vacinação/veterinária
13.
J Infect Dis ; 220(6): 956-960, 2019 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-31056697

RESUMO

In this post-hoc analysis of midnasal pneumococcal carriage in a community-based, randomized prenatal influenza vaccination trial in Nepal with weekly infant respiratory illness surveillance, 457 of 605 (75.5%) infants with influenza, respiratory syncytial virus (RSV), or human metapneumovirus (hMPV) illness had pneumococcus detected. Pneumococcal carriage did not impact rates of lower respiratory tract disease for these 3 viruses. Influenza-positive infants born to mothers given influenza vaccine had lower pneumococcal carriage rates compared to influenza-positive infants born to mothers receiving placebo (58.1% versus 71.6%, P = 0.03). Maternal influenza immunization may impact infant acquisition of pneumococcus during influenza infection. Clinical Trials Registration. NCT01034254.


Assuntos
Vacinas contra Influenza , Influenza Humana/epidemiologia , Infecções por Paramyxoviridae/epidemiologia , Infecções Pneumocócicas/epidemiologia , Infecções por Vírus Respiratório Sincicial/epidemiologia , Vacinação , Feminino , Humanos , Lactente , Recém-Nascido , Influenza Humana/prevenção & controle , Metapneumovirus , Mães , Nepal , Infecções por Paramyxoviridae/prevenção & controle , Infecções Pneumocócicas/prevenção & controle , Gravidez , Complicações Infecciosas na Gravidez/prevenção & controle , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vírus Sincicial Respiratório Humano , Infecções Respiratórias/epidemiologia , Infecções Respiratórias/prevenção & controle , Infecções Respiratórias/virologia , Streptococcus pneumoniae
14.
Vaccine ; 37(21): 2765-2767, 2019 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-31003913

RESUMO

Since late '80 s Avian metapneumovirus subtype A causes sufficient disease in Europe for commercial companies to have started developing live attenuated vaccines. Here, two of those vaccines were fully consensus sequenced alongside their progenitor field strain (#8544). Sequences comparison shows that the attenuation of field strain #8544 was associated with no common substitutions between the two derived vaccines. This finding suggests that the attenuation of field viruses via serial passage on cell cultures or tissues is the result of a random process, rather than a mechanism aiming to achieve a specific sequence. Furthermore, field vaccination strategies would greatly benefit by the unambiguous vaccine markers identified in this study, enabling a prompt and confident vaccines detection.


Assuntos
Metapneumovirus/imunologia , Metapneumovirus/patogenicidade , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Vacinação/efeitos adversos , Vacinas Atenuadas/uso terapêutico , Vacinas Virais/uso terapêutico , Animais , Metapneumovirus/genética , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/prevenção & controle , RNA Viral/genética , Perus
15.
Sci Rep ; 8(1): 10425, 2018 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-29992955

RESUMO

Paramyxoviridae, a large family of enveloped viruses harboring a nonsegmented negative-sense RNA genome, include important human pathogens as measles, mumps, respiratory syncytial virus (RSV), parainfluenza viruses, and henipaviruses, which cause some of the deadliest emerging zoonoses. There is no effective antiviral chemotherapy for most of these pathogens. Paramyxoviruses evolved a sophisticated membrane-fusion machine consisting of receptor-binding proteins and the fusion F-protein, critical for virus infectivity. Herein we identify the antiprotozoal/antimicrobial nitazoxanide as a potential anti-paramyxovirus drug targeting the F-protein. We show that nitazoxanide and its circulating-metabolite tizoxanide act at post-entry level by provoking Sendai virus and RSV F-protein aggregate formation, halting F-trafficking to the host plasma membrane. F-protein folding depends on ER-resident glycoprotein-specific thiol-oxidoreductase ERp57 for correct disulfide-bond architecture. We found that tizoxanide behaves as an ERp57 non-competitive inhibitor; the putative drug binding-site was located at the ERp57-b/b' non-catalytic domains interface. ERp57-silencing mimicked thiazolide-induced F-protein alterations, suggesting an important role of this foldase in thiazolides anti-paramyxovirus activity. Nitazoxanide is used in the clinic as a safe and effective antiprotozoal/antimicrobial drug; its antiviral activity was shown in patients infected with hepatitis-C virus, rotavirus and influenza viruses. Our results now suggest that nitazoxanide may be effective also against paramyxovirus infection.


Assuntos
Infecções por Paramyxoviridae/tratamento farmacológico , Paramyxoviridae/fisiologia , Tiazóis/farmacologia , Replicação Viral/efeitos dos fármacos , Células A549 , Animais , Sítios de Ligação , Humanos , Nitrocompostos , Oxirredutases/metabolismo , Paramyxoviridae/efeitos dos fármacos , Infecções por Paramyxoviridae/prevenção & controle , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Isomerases de Dissulfetos de Proteínas/química , Dobramento de Proteína/efeitos dos fármacos , Transporte Proteico , Tiazóis/metabolismo , Proteínas Virais de Fusão/metabolismo
16.
Viral Immunol ; 31(2): 133-141, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29323621

RESUMO

Human parainfluenza viruses (family Paramyxoviridae), human metapneumovirus, and respiratory syncytial virus (family Pneumoviridae) infect most infants and children within the first few years of life and are the etiologic agents for many serious acute respiratory illnesses. These virus infections are also associated with long-term diseases that impact quality of life, including asthma. Despite over a half-century of vaccine research, development, and clinical trials, no vaccine has been licensed to date for the paramyxoviruses or pneumoviruses for the youngest infants. In this study, we describe the recent reclassification of paramyxoviruses and pneumoviruses into distinct families by the International Committee on the Taxonomy of Viruses. We also discuss some past unsuccessful vaccine trials and some currently preferred vaccine strategies. Finally, we discuss hurdles that must be overcome to support successful respiratory virus vaccine development for the youngest children.


Assuntos
Descoberta de Drogas/tendências , Infecções por Paramyxoviridae/prevenção & controle , Paramyxovirinae/imunologia , Pneumovirinae/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas Virais/imunologia , Vacinas Virais/isolamento & purificação , Animais , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Humanos , Infecções por Paramyxoviridae/epidemiologia , Paramyxovirinae/classificação , Pneumovirinae/classificação , Infecções por Vírus Respiratório Sincicial/epidemiologia
17.
Viral Immunol ; 31(4): 306-314, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29373084

RESUMO

The human metapneumovirus (hMPV) is the second leading cause globally of acute infection of the respiratory tract in children, infecting the upper and lower airways. The hMPV may induce an inappropriate Th2-type immune response, which causes severe pulmonary inflammation, leading to the obstruction of airways. Despite its severe epidemiological relevance, no vaccines are currently available for the prevention of hMPV-induced illness. In this investigation, we demonstrated that immunization of mice with the recombinant hMPV nucleoprotein (hMPV-N) mixed with the AbISCO-100 adjuvant reduced viral replication in lungs following challenge with the virus. We found that immunized mice had reduced weight loss, decreased granulocytes in the lung, an increased level of specific nucleoprotein antibodies of IgG1 and IgG2a-isotypes, and a local profile of Th1/Th17-type cytokines. Our results suggest that immunization with the hMPV-N and the AbISCO-100 adjuvant induces a reduction of viral infection and could be considered for the development of an hMPV vaccine.


Assuntos
Imunização , Metapneumovirus/imunologia , Nucleoproteínas/administração & dosagem , Infecções por Paramyxoviridae/imunologia , Vacinas Virais/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/classificação , Citocinas/análise , Células Dendríticas/classificação , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Granulócitos , Humanos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Nucleoproteínas/genética , Nucleoproteínas/isolamento & purificação , Infecções por Paramyxoviridae/prevenção & controle , Pneumonia/virologia , RNA Viral/análise , Vacinas Virais/farmacologia , Redução de Peso
18.
J Pediatric Infect Dis Soc ; 7(1): 86-89, 2018 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-28444226

RESUMO

We conducted a phase I clinical trial of an experimental live attenuated recombinant human metapneumovirus (HMPV) vaccine (rHMPV-Pa) sequentially in adults, HMPV-seropositive children, and HMPV-seronegative children, the target population for vaccination. rHMPV-Pa was appropriately restricted in replication in adults and HMPV-seropositive children but was overattenuated for HMPV-seronegative children.


Assuntos
Metapneumovirus/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Vacinas Virais/uso terapêutico , Adulto , Anticorpos Antivirais/imunologia , Pré-Escolar , Método Duplo-Cego , Humanos , Lactente , Infecções por Paramyxoviridae/terapia , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/uso terapêutico , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico , Vacinas Virais/imunologia
19.
Virology ; 509: 60-66, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28605636

RESUMO

Human metapneumovirus (hMPV) infections pose a serious health risk to young children, particularly in cases of premature birth. No licensed vaccine exists and there is no standard treatment for hMPV infections apart from supportive hospital care. We describe the production of a Sendai virus (SeV) recombinant that carries a gene for a truncated hMPV fusion (F) protein (SeV-MPV-Ft). The vaccine induces binding and neutralizing antibody responses toward hMPV and protection against challenge with hMPV in a cotton rat system. Results encourage advanced development of SeV-MPV-Ft to prevent the morbidity and mortality caused by hMPV infections in young children.


Assuntos
Antígenos Virais/imunologia , Portadores de Fármacos , Metapneumovirus/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Vírus Sendai/genética , Proteínas Virais de Fusão/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Antígenos Virais/genética , Modelos Animais de Doenças , Metapneumovirus/genética , Infecções por Paramyxoviridae/imunologia , Sigmodontinae , Resultado do Tratamento , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas Virais de Fusão/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
20.
Sci Rep ; 7(1): 4025, 2017 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-28642611

RESUMO

Avian metapneumovirus (AMPV) infects the respiratory and reproductive tracts of domestic poultry, resulting in substantial economic losses for producers. Live attenuated vaccines appear to be the most effective in countries where the disease is prevalent. However, reversion to virulence has been demonstrated in several studies. Therefore, the development of a stable and safe next generation vaccine against the AMPV disease is needed. In the present study, we generated a recombinant Newcastle disease virus (NDV) vectoring the fusion (F) protein and glycoprotein (G) genes of AMPV subtype-C (AMPV-C) as a bivalent vaccine candidate using reverse genetics technology. The recombinant virus, rLS/AMPV-C F&G, was slightly attenuated in vivo, yet maintained similar characteristics in vitro when compared to the parental LaSota virus. Vaccination of turkeys with rLS/AMPV-C F&G induced both AMPV-C and NDV-specific antibody responses, and provided significant protection against pathogenic AMPV-C challenge and complete protection against velogenic NDV challenge. These results suggest that the rLS/AMPV-C F&G recombinant virus is a safe and effective bivalent vaccine candidate and that the expression of both F and G proteins of AMPV-C induces a protective response against the AMPV-C disease.


Assuntos
Vetores Genéticos/genética , Metapneumovirus/genética , Metapneumovirus/imunologia , Vírus da Doença de Newcastle/genética , Proteínas do Envelope Viral/genética , Proteínas Virais de Fusão/genética , Vacinas Virais/genética , Animais , Anticorpos Antivirais/imunologia , Expressão Gênica , Ordem dos Genes , Vetores Genéticos/imunologia , Imunidade Humoral , Imunização , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Infecções por Paramyxoviridae/virologia , Doenças das Aves Domésticas/prevenção & controle , Perus , Proteínas do Envelope Viral/imunologia , Proteínas Virais de Fusão/imunologia , Vacinas Virais/imunologia , Eliminação de Partículas Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA