Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.198
Filtrar
1.
Front Immunol ; 15: 1382318, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38646538

RESUMO

The respiratory syncytial virus (RSV) is a leading cause of acute lower respiratory tract infections associated with numerous hospitalizations. Recently, intramuscular (i.m.) vaccines against RSV have been approved for elderly and pregnant women. Noninvasive mucosal vaccination, e.g., by inhalation, offers an alternative against respiratory pathogens like RSV. Effective mucosal vaccines induce local immune responses, potentially resulting in the efficient and fast elimination of respiratory viruses after natural infection. To investigate this immune response to an RSV challenge, low-energy electron inactivated RSV (LEEI-RSV) was formulated with phosphatidylcholine-liposomes (PC-LEEI-RSV) or 1,2-dioleoyl-3-trimethylammonium-propane and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DD-LEEI-RSV) for vaccination of mice intranasally. As controls, LEEI-RSV and formalin-inactivated-RSV (FI-RSV) were used via i.m. vaccination. The RSV-specific immunogenicity of the different vaccines and their protective efficacy were analyzed. RSV-specific IgA antibodies and a statistically significant reduction in viral load upon challenge were detected in mucosal DD-LEEI-RSV-vaccinated animals. Alhydrogel-adjuvanted LEEI-RSV i.m. showed a Th2-bias with enhanced IgE, eosinophils, and lung histopathology comparable to FI-RSV. These effects were absent when applying the mucosal vaccines highlighting the potential of DD-LEEI-RSV as an RSV vaccine candidate and the improved performance of this mucosal vaccine candidate.


Assuntos
Anticorpos Antivirais , Imunidade nas Mucosas , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial , Vacinas contra Vírus Sincicial Respiratório , Células Th2 , Vacinas de Produtos Inativados , Animais , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vacinas contra Vírus Sincicial Respiratório/administração & dosagem , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Infecções por Vírus Respiratório Sincicial/imunologia , Camundongos , Vacinas de Produtos Inativados/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Feminino , Células Th2/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Imunização , Vírus Sincicial Respiratório Humano/imunologia , Vacinação/métodos , Vírus Sinciciais Respiratórios/imunologia , Carga Viral , Imunoglobulina A/imunologia
2.
Arch. bronconeumol. (Ed. impr.) ; 60(3): 161-170, Mar. 2024. tab, ilus
Artigo em Inglês | IBECS | ID: ibc-231100

RESUMO

Respiratory syncytial virus (RSV) is a major cause of respiratory tract infections in adults, particularly older adults and those with underlying medical conditions. Vaccination has emerged as a potential key strategy to prevent RSV-related morbidity and mortality. This Neumoexperts Prevention (NEP) Group scientific paper aims to provide an evidence-based positioning and RSV vaccination recommendations for adult patients. We review the current literature on RSV burden and vaccine development and availability, emphasising the importance of vaccination in the adult population. According to our interpretation of the data, RSV vaccines should be part of the adult immunisation programme, and an age-based strategy should be preferred over targeting high-risk groups. The effectiveness and efficiency of this practice will depend on the duration of protection and the need for annual or more spaced doses. Our recommendations should help healthcare professionals formulate guidelines and implement effective vaccination programmes for adult patients at risk of RSV infection now that specific vaccines are available.(AU)


Assuntos
Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Vírus Sinciciais Respiratórios/imunologia , Vacinação , Prevenção de Doenças , Pneumopatias/prevenção & controle , Pneumopatias/imunologia , Programas de Imunização
3.
MMWR recomm. rep ; 72(34): 1-4, 20230825. tab
Artigo em Inglês | BIGG - guias GRADE | ID: biblio-1512312

RESUMO

A Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) review of the evidence for benefits and harms for the long-acting monoclonal antibody nirsevimab, (Beyfortus, Sanofi and AstraZeneca) for prevention of respiratory syncytial virus (RSV))-associated lower respiratory tract infection (LRTI) in children <24 months who are at increased risk of severe disease entering their second RSV season was presented to the Advisory Committee on Immunization Practices (ACIP) on February 23, 2023. GRADE evidence type indicates the certainty in estimates from the available body of evidence. Evidence certainty ranges from high certainty to very low certainty


Assuntos
Humanos , Recém-Nascido , Lactente , Vírus Sinciciais Respiratórios/imunologia , Anticorpos Monoclonais/uso terapêutico
4.
J Biol Chem ; 299(8): 105028, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37423306

RESUMO

As part of the antiviral response, cells activate the expressions of type I interferons (IFNs) and proinflammatory mediators to control viral spreading. Viral infections can impact DNA integrity; however, how DNA damage repair coordinates antiviral response remains elusive. Here we report Nei-like DNA glycosylase 2 (NEIL2), a transcription-coupled DNA repair protein, actively recognizes the oxidative DNA substrates induced by respiratory syncytial virus (RSV) infection to set the threshold of IFN-ß expression. Our results show that NEIL2 antagonizes nuclear factor κB (NF-κB) acting on the IFN-ß promoter early after infection, thus limiting gene expression amplified by type I IFNs. Mice lacking Neil2 are far more susceptible to RSV-induced illness with an exuberant expression of proinflammatory genes and tissue damage, and the administration of NEIL2 protein into the airway corrected these defects. These results suggest a safeguarding function of NEIL2 in controlling IFN-ß levels against RSV infection. Due to the short- and long-term side effects of type I IFNs applied in antiviral therapy, NEIL2 may provide an alternative not only for ensuring genome fidelity but also for controlling immune responses.


Assuntos
DNA Glicosilases , Interferon beta , Infecções por Vírus Respiratório Sincicial , Vírus Sinciciais Respiratórios , Animais , Camundongos , DNA , DNA Glicosilases/genética , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Interferon beta/genética , Infecções por Vírus Respiratório Sincicial/genética , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/imunologia
5.
MMWR recomm. rep ; 79(29): 1-6, 20230721. tab
Artigo em Inglês | BIGG - guias GRADE | ID: biblio-1512316

RESUMO

A Grading of Recommendations, Assessment, Development and Evaluation (GRADE) review of the evidence for benefits and harms for GSK Respiratory Syncytial Virus (RSV) PreF3 vaccine was presented to the Advisory Committee on Immunization Practices (ACIP) on June 21, 2023. GRADE evidence type indicates the certainty in estimates from the available body of evidence. Evidence certainty ranges from high certainty to very low certainty. The policy questions were, "Should vaccination with GSK RSVPreF3 vaccine (120µg antigen + AS01E adjuvant, 1 dose administered intramuscularly [IM]), rather than no vaccine, be recommended in persons aged ≥65 years?" and "Should vaccination with GSK RSVPreF3 vaccine (120µg antigen + AS01E adjuvant, 1 dose IM), rather than no vaccine, be recommended in persons aged 60­64 years?" The benefits chosen by the ACIP RSV Vaccines Work Group (Work Group) as critical or important to policy decisions were prevention of RSV lower respiratory tract illness/disease (LRTI/LRTD) (critical), medically attended RSV LRTI/LRTD (critical), hospitalization for RSV respiratory illness (important), severe RSV respiratory illness requiring supplemental oxygen (O2) or other respiratory support (important), and death due to RSV respiratory illness (important). The harms chosen by the Work Group as critical or important to policy decisions were serious adverse events (critical), inflammatory neurologic events* (important) and reactogenicity grade ≥3 (important). A systematic review of evidence on the efficacy and safety of GSK RSVPreF3 vaccine among persons aged 60 years and older was conducted. The quality of evidence from one Phase 3 randomized controlled trial (RCT) and one Phase 1/2 RCT were assessed using the GRADE approach [2-4]. Efficacy findings were based on analyses of data collected during May 2021­March 2023, which included two complete RSV seasons for Northern Hemisphere participants and one complete RSV season for Southern Hemisphere participants. A lower risk of RSV LRTD† was observed with vaccination compared to placebo (incident rate ratio [IRR] 0.254, 95% confidence interval [CI]: 0.165, 0.379, evidence certainty: moderate), corresponding to a vaccine efficacy of 74.6% (95% CI: 62.1%, 83.5%)§. A lower risk of medically attended RSV LRTD¶ was also observed (IRR 0.225; 95% CI: 0.110, 0.421; evidence certainty: moderate), corresponding to a vaccine efficacy of 77.5% (95% CI: 57.9%, 89.0%).** The trial was not powered to detect a lower risk of hospitalization for RSV respiratory illness or severe RSV respiratory illness requiring supplemental oxygen or other respiratory support (for both outcomes IRR 0.236; 95% CI: 0.005, 2.112; evidence certainty: very low), corresponding to a vaccine efficacy for both outcomes of 76.4% (95% CI: -111%, 99.5%). No deaths due to RSV respiratory illness were identified among vaccine recipients or placebo recipients. In terms of harms, the pooled available data from the Phase 3 and Phase 1/2 RCTs indicated that serious adverse events (SAEs)†† were balanced between participants in the vaccine and placebo arms (risk ratio [RR] 1.019; 95% CI: 0.908, 1.145; evidence certainty: high). Reactogenicity grade ≥3§§ was associated with vaccination (RR 4.099; 95% CI: 1.989, 8.446; evidence certainty: high), with 3.8% of vaccine recipients and 0.9% of placebo recipients reporting any grade ≥3 local or systemic reactions following injection. No inflammatory neurologic events were observed within 42 days after injection in either placebo-controlled trial. However, inflammatory neurologic events were observed in other trials not included in the GRADE assessment due to lack of an unvaccinated comparator: one event of Guillain-Barré syndrome (GBS) reported within 42 days after vaccination in a recipient of the investigational vaccine in an open label trial without a placebo arm and two events of acute disseminated encephalomyelitis (ADEM) reported within 42 days after coadministration of the investigational vaccine with standard dose seasonal influenza vaccine in a coadministration study (for one of these ADEM cases the investigator revised the diagnosis to hypoglycemia and dementia in June of 2023).


Assuntos
Humanos , Adulto , Pessoa de Meia-Idade , Idoso , Vírus Sinciciais Respiratórios/imunologia , Vacinas contra Influenza/uso terapêutico , Desenvolvimento de Vacinas
6.
PLoS Pathog ; 18(2): e1010272, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35108347

RESUMO

Respiratory syncytial virus (RSV) can cause bronchiolitis and viral pneumonia in young children and the elderly. Lack of vaccines and recurrence of RSV infection indicate the difficulty in eliciting protective memory immune responses. Tissue resident memory T cells (TRM) can confer protection from pathogen re-infection and, in human experimental RSV infection, the presence of lung CD8+ TRM cells correlates with a better outcome. However, the requirements for generating and maintaining lung TRM cells during RSV infection are not fully understood. Here, we use mouse models to assess the impact of innate immune response determinants in the generation and subsequent expansion of the TRM cell pool during RSV infection. We show that CD8+ TRM cells expand independently from systemic CD8+ T cells after RSV re-infection. Re-infected MAVS and MyD88/TRIF deficient mice, lacking key components involved in innate immune recognition of RSV and induction of type I interferons (IFN-α/ß), display impaired expansion of CD8+ TRM cells and reduction in antigen specific production of granzyme B and IFN-γ. IFN-α treatment of MAVS deficient mice during primary RSV infection restored TRM cell expansion upon re-challenge but failed to recover TRM cell functionality. Our data reveal how innate immunity, including the axis controlling type I IFN induction, instructs and regulates CD8+ TRM cell responses to RSV infection, suggesting possible mechanisms for therapeutic intervention.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Linfócitos T CD8-Positivos/imunologia , Interferon Tipo I/imunologia , Células T de Memória/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Animais , Granzimas/imunologia , Granzimas/metabolismo , Imunidade Inata , Memória Imunológica , Interferon Tipo I/metabolismo , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Vírus Respiratório Sincicial/virologia , Transdução de Sinais
7.
J Immunol ; 208(3): 720-731, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35022269

RESUMO

Respiratory infections are a leading cause of morbidity and mortality. The presence of multiple heterologous virus infections is routinely observed in a subset of individuals screened for the presence of respiratory viruses. However, the impact overlapping infections has on disease severity and the host immune response is not well understood. Respiratory syncytial virus (RSV) and influenza A virus (IAV) are two of the most common respiratory infections observed in hospitalized patients, particularly in the very young and aged populations. In this study, we examined how the order in which BALB/c mice were infected with both RSV and IAV impacts disease severity. RSV infection prior to an IAV infection was associated with decreased weight loss and increased survival as compared with IAV infection alone. In contrast, IAV infection prior to an RSV infection was associated with similar morbidity and mortality as compared with an IAV infection alone. Our results suggest that the order in which viral infections are acquired plays a critical role in the outcome of disease severity and the host immune response.


Assuntos
Vírus da Influenza A/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Interferência Viral/fisiologia , Animais , Anticorpos Antivirais/imunologia , Linfócitos T CD8-Positivos/imunologia , Coinfecção/imunologia , Coinfecção/virologia , Citocinas/imunologia , Feminino , Interferon Tipo I/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/prevenção & controle
8.
Am J Respir Cell Mol Biol ; 66(2): 206-222, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34731594

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected more than 180 million people since the onset of the pandemic. Despite similar viral load and infectivity rates between children and adults, children rarely develop severe illness. Differences in the host response to the virus at the primary infection site are among the mechanisms proposed to account for this disparity. Our objective was to investigate the host response to SARS-CoV-2 in the nasal mucosa in children and adults and compare it with the host response to respiratory syncytial virus (RSV) and influenza virus. We analyzed clinical outcomes and gene expression in the nasal mucosa of 36 children with SARS-CoV-2, 24 children with RSV, 9 children with influenza virus, 16 adults with SARS-CoV-2, and 7 healthy pediatric and 13 healthy adult controls. In both children and adults, infection with SARS-CoV-2 led to an IFN response in the nasal mucosa. The magnitude of the IFN response correlated with the abundance of viral reads, not the severity of illness, and was comparable between children and adults infected with SARS-CoV-2 and children with severe RSV infection. Expression of ACE2 and TMPRSS2 did not correlate with age or presence of viral infection. SARS-CoV-2-infected adults had increased expression of genes involved in neutrophil activation and T-cell receptor signaling pathways compared with SARS-CoV-2-infected children, despite similar severity of illness and viral reads. Age-related differences in the immune response to SARS-CoV-2 may place adults at increased risk of developing severe illness.


Assuntos
Envelhecimento/imunologia , COVID-19/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade nas Mucosas , Mucosa Nasal/imunologia , SARS-CoV-2/imunologia , Adolescente , Fatores Etários , Enzima de Conversão de Angiotensina 2/imunologia , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Mucosa Nasal/virologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Serina Endopeptidases/imunologia
9.
J Leukoc Biol ; 111(2): 379-389, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-33866604

RESUMO

The activation of dendritic cells (DC) during respiratory viral infections is central to directing the immune response and the pathologic outcome. In these studies, the effect of RSV infection on development of ER stress responses and the impact on innate immunity was examined. The upregulation of ER stress was closely associated with the PERK pathway through the upregulation of CHOP in RSV infected DC. The inhibition of PERK corresponded with decreased EIF2a phosphorylation but had no significant effect on Nrf2 in DC, two primary pathways regulated by PERK. Subsequent studies identified that by blocking PERK activity in infected DC an altered ER stress response and innate cytokine profile was observed with the upregulation of IFNß and IL-12, coincident to the down regulation of IL-1ß. When mitochondria respiration was assessed in PERK deficient DC there were increased dysfunctional mitochondria after RSV infection that resulted in reduced oxygen consumption rates (OCR) and ATP production indicating altered cellular metabolism. Use of a CD11c targeted genetic deleted murine model, RSV infection was characterized by reduced inflammation and diminished mucus staining as well as reduced mucus-associated gene gob5 expression. The assessment of the cytokine responses showed decreased IL-13 and IL-17 along with diminished IL-1ß in the lungs of PERK deficient infected mice. When PERK-deficient animals were assessed in parallel for lung leukocyte numbers, animals displayed significantly reduced myeloid and activated CD4 and CD8 T cell numbers. Thus, the PERK activation pathway may provide a rational target for altering the severe outcome of an RSV infection through modifying immune responses.


Assuntos
Células Dendríticas/imunologia , Estresse do Retículo Endoplasmático , Imunidade Inata , Inflamação/patologia , Infecções por Vírus Respiratório Sincicial/patologia , Vírus Sinciciais Respiratórios/imunologia , eIF-2 Quinase/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Citocinas/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Consumo de Oxigênio , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/metabolismo , eIF-2 Quinase/genética
11.
J Immunol ; 207(10): 2589-2597, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34625522

RESUMO

Respiratory syncytial virus (RSV) infection in infancy is associated with increased risk of asthma, except in those with allergic disease at the time of infection. Using house dust mite allergen, we examined the effect of pre-existing atopy on postviral airway disease using Sendai virus in mice, which models RSV infection in humans. Sendai virus drives postviral airway disease in nonatopic mice; however, pre-existing atopy protected against the development of airway disease. This protection depended upon neutrophils, as depletion of neutrophils at the time of infection restored the susceptibility of atopic mice to postviral airway disease. Associated with development of atopy was an increase in polymorphonuclear neutrophil-dendritic cell hybrid cells that develop in Th2 conditions and demonstrated increased viral uptake. Systemic inhibition of IL-4 reversed atopic protection against postviral airway disease, suggesting that increased virus uptake by neutrophils was IL-4 dependent. Finally, human neutrophils from atopic donors were able to reduce RSV infection of human airway epithelial cells in vitro, suggesting these findings could apply to the human. Collectively our data support the idea that pre-existing atopy derives a protective neutrophil response via potential interaction with IL-4, preventing development of postviral airway disease.


Assuntos
Hipersensibilidade Imediata/imunologia , Neutrófilos/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Respirovirus/imunologia , Animais , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Vírus Sinciciais Respiratórios/imunologia , Vírus Sendai/imunologia
12.
Elife ; 102021 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-34586067

RESUMO

The complement system is a critical host defense against infection, playing a protective role that can also enhance disease if dysregulated. Although many consequences of complement activation during viral infection are well established, mechanisms that determine the extent to which viruses activate complement remain elusive. Here, we investigate complement activation by human respiratory syncytial virus (RSV), a filamentous respiratory pathogen that causes significant morbidity and mortality. By engineering a strain of RSV harboring tags on the surface glycoproteins F and G, we are able to monitor opsonization of single RSV particles using fluorescence microscopy. These experiments reveal an antigenic hierarchy, where antibodies that bind toward the apex of F in either the pre- or postfusion conformation activate the classical pathway whereas other antibodies do not. Additionally, we identify an important role for virus morphology in complement activation: as viral filaments age, they undergo a morphological transformation which lowers the threshold for complement deposition through changes in surface curvature. Collectively, these results identify antigenic and biophysical characteristics of virus particles that contribute to the formation of viral immune complexes, and suggest models for how these factors may shape disease severity and adaptive immune responses to RSV.


Assuntos
Proteínas do Sistema Complemento/metabolismo , Vírus Sinciciais Respiratórios/metabolismo , Animais , Anticorpos Antivirais/imunologia , Linhagem Celular , Ativação do Complemento , Humanos , Modelos Biológicos , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/imunologia
13.
Respirology ; 26(11): 1049-1059, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34472161

RESUMO

BACKGROUND AND OBJECTIVE: Influenza virus (FLU), rhinovirus (RV) and respiratory syncytial virus (RSV) are the most common acute respiratory infections worldwide. Infection can cause severe health outcomes, while therapeutic options are limited, primarily relieving symptoms without attenuating the development of lesions or impaired lung function. We therefore examined the inflammatory response to these infections with the intent to identify common components that are critical drivers of immunopathogenesis and thus represent potential therapeutic targets. METHODS: BALB/c mice were infected with FLU, RV or RSV, and lung function, airway inflammation and immunohistopathology were measured over a 10-day period. Anti-IL-17A mAb was administered to determine the impact of attenuating this cytokine's function on the development and severity of disease. RESULTS: All three viruses induced severe airway constriction and inflammation at 2 days post-infection (dpi). However, only FLU induced prolonged inflammation till 10 dpi. Increased IL-17A expression was correlated with the alterations in lung function and its persistence. Neutralization of IL-17A did not affect the viral replication but led to the resolution of airway hyperresponsiveness. Furthermore, anti-IL-17A treatment resulted in reduced infiltration of neutrophils (in RV- and FLU-infected mice at 2 dpi) and lymphocytes (in RSV-infected mice at 2 dpi and FLU-infected mice at 10 dpi), and attenuated the severity of immunopathology. CONCLUSION: IL-17A is a common pathogenic molecule regulating disease induced by three prevalent respiratory viruses. Targeting the IL-17A pathway may provide a unified approach to the treatment of these respiratory infections alleviating both inflammation-induced lesions and difficulties in breathing.


Assuntos
Interleucina-17/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Picornaviridae/imunologia , Infecções por Vírus Respiratório Sincicial , Animais , Pulmão/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Orthomyxoviridae , Vírus Sinciciais Respiratórios/imunologia , Rhinovirus
14.
Front Immunol ; 12: 704427, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34489955

RESUMO

Viral pneumonias are a major cause of morbidity and mortality, owing in part to dysregulated excessive lung inflammation, and therapies to modulate host responses to viral lung injury are urgently needed. Protectin conjugates in tissue regeneration 1 (PCTR1) and protectin D1 (PD1) are specialized pro-resolving mediators (SPMs) whose roles in viral pneumonia are of interest. In a mouse model of Respiratory Syncytial Virus (RSV) pneumonia, intranasal PCTR1 and PD1 each decreased RSV genomic viral load in lung tissue when given after RSV infection. Concurrent with enhanced viral clearance, PCTR1 administration post-infection, decreased eosinophils, neutrophils, and NK cells, including NKG2D+ activated NK cells, in the lung. Intranasal PD1 administration post-infection decreased lung eosinophils and Il-13 expression. PCTR1 increased lung expression of cathelicidin anti-microbial peptide and decreased interferon-gamma production by lung CD4+ T cells. PCTR1 and PD1 each increased interferon-lambda expression in human bronchial epithelial cells in vitro and attenuated RSV-induced suppression of interferon-lambda in mouse lung in vivo. Liquid chromatography coupled with tandem mass spectrometry of RSV-infected and untreated mouse lungs demonstrated endogenous PCTR1 and PD1 that decreased early in the time course while cysteinyl-leukotrienes (cys-LTs) increased during early infection. As RSV infection resolved, PCTR1 and PD1 increased and cys-LTs decreased to pre-infection levels. Together, these results indicate that PCTR1 and PD1 are each regulated during RSV pneumonia, with overlapping and distinct mechanisms for PCTR1 and PD1 during the resolution of viral infection and its associated inflammation.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Pulmão/imunologia , Pneumonia Viral , Infecções por Vírus Respiratório Sincicial , Vírus Sinciciais Respiratórios/imunologia , Administração Intranasal , Animais , Inflamação/imunologia , Inflamação/prevenção & controle , Pulmão/virologia , Masculino , Camundongos , Pneumonia Viral/imunologia , Pneumonia Viral/prevenção & controle , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Carga Viral/efeitos dos fármacos
15.
Sci Rep ; 11(1): 15694, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34344952

RESUMO

Respiratory syncytial virus (RSV) infection is a common cause of hospitalisation in infants and the elderly. Palivizumab prophylaxis is the only approved treatment modality but is costly and only offered to select vulnerable populations. Here, we investigated gene delivery approaches via recombinant adeno-associated virus (rAAV2/8) and simian immunodeficiency virus (rSIV.F/HN) vectors to achieve sustained in vivo production of palivizumab in a murine model. Delivery of palivizumab-expressing vectors 28 days prior to RSV challenge resulted in complete protection from RSV-induced weight loss. This approach offers prophylaxis against RSV infection, allowing for wider use and reduction in treatment costs in vulnerable populations.


Assuntos
Anticorpos Monoclonais Humanizados/genética , Expressão Gênica , Terapia Genética , Palivizumab/genética , Infecções por Vírus Respiratório Sincicial/terapia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios , Animais , Dependovirus/genética , Modelos Animais de Doenças , Feminino , Técnicas de Transferência de Genes , Engenharia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Injeções Intramusculares , Lentivirus/genética , Camundongos , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Transdução Genética , Resultado do Tratamento
16.
J Med Virol ; 93(11): 6116-6123, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34375002

RESUMO

Virus invasion activates the host's innate immune response, inducing the production of numerous cytokines and interferons to eliminate pathogens. Except for viral DNA/RNA, viral proteins are also targets of pattern recognition receptors. Membrane-bound receptors such as Toll-like receptor (TLR)1, TLR2, TLR4, TLR6, and TLR10 relate to the recognition of viral proteins. Distinct TLRs perform both protective and detrimental roles for a specific virus. Here, we review viral proteins serving as pathogen-associated molecular patterns and their corresponding TLRs. These viruses are all enveloped, including respiratory syncytial virus, hepatitis C virus, measles virus, herpesvirus human immunodeficiency virus, and coronavirus, and can encode proteins to activate innate immunity in a TLR-dependent way. The TLR-viral protein relationship plays an important role in innate immunity activation. A detailed understanding of their pathways contributes to a novel direction for vaccine development.


Assuntos
Imunidade Inata , Moléculas com Motivos Associados a Patógenos/metabolismo , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Proteínas Virais/metabolismo , Viroses/imunologia , Vírus/imunologia , Animais , HIV/imunologia , HIV/metabolismo , HIV/patogenicidade , Hepacivirus/imunologia , Hepacivirus/metabolismo , Hepacivirus/patogenicidade , Herpesviridae/imunologia , Herpesviridae/metabolismo , Herpesviridae/patogenicidade , Humanos , Vírus do Sarampo/imunologia , Vírus do Sarampo/metabolismo , Vírus do Sarampo/patogenicidade , Moléculas com Motivos Associados a Patógenos/química , Vírus Sinciciais Respiratórios/imunologia , Vírus Sinciciais Respiratórios/metabolismo , Vírus Sinciciais Respiratórios/patogenicidade , SARS-CoV-2/imunologia , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Proteínas Virais/química , Viroses/virologia , Vírus/metabolismo , Vírus/patogenicidade
17.
Virology ; 563: 28-37, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34411809

RESUMO

Respiratory syncytial virus (RSV) infection is not only a childhood disease, but also a serious health risk for the elderly. We investigated in cotton rats how age affected viral clearance, immune responses, and whether pharmacological intervention was beneficial. Our results demonstrated that in geriatric animals, virus grew to similar titers, but with delayed clearance, compared to adult animals. After primary infection with RSV, geriatric animals were susceptible to secondary infection and results indicated a defective humoral immune response. Depletion of cytotoxic T lymphocytes (CTL) during primary infection delayed clearance, indicating the necessary role of CTL. Pharmacological intervention through nonsteroidal anti-inflammatory ibuprofen resulted in faster viral clearance and complete protection after immunization. In addition, the CTL response in the presence of ibuprofen seemed to be restored. It appears that in geriatric animals, immune functions are not as effective as in adult animals and that anti-inflammatory therapy may restore effective immune function.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Ibuprofeno/uso terapêutico , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Envelhecimento , Animais , Feminino , Imunidade Humoral , Masculino , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/imunologia , Sigmodontinae , Linfócitos T Citotóxicos
20.
Proc Natl Acad Sci U S A ; 118(14)2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33811145

RESUMO

Human respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory infection in children under 5 y of age. In the absence of a safe and effective vaccine and with limited options for therapeutic interventions, uncontrolled epidemics of RSV occur annually worldwide. Existing RSV reverse genetics systems have been predominantly based on older laboratory-adapted strains such as A2 or Long. These strains are not representative of currently circulating genotypes and have a convoluted passage history, complicating their use in studies on molecular determinants of viral pathogenesis and intervention strategies. In this study, we have generated reverse genetics systems for clinical isolates of RSV-A (ON1, 0594 strain) and RSV-B (BA9, 9671 strain) in which the full-length complementary DNA (cDNA) copy of the viral antigenome is cloned into a bacterial artificial chromosome (BAC). Additional recombinant (r) RSVs were rescued expressing enhanced green fluorescent protein (EGFP), mScarlet, or NanoLuc luciferase from an additional transcription unit inserted between the P and M genes. Mutations in antigenic site II of the F protein conferring escape from palivizumab neutralization (K272E, K272Q, S275L) were investigated using quantitative cell-fusion assays and rRSVs via the use of BAC recombineering protocols. These mutations enabled RSV-A and -B to escape palivizumab neutralization but had differential impacts on cell-to-cell fusion, as the S275L mutation resulted in an almost-complete ablation of syncytium formation. These reverse genetics systems will facilitate future cross-validation efficacy studies of novel RSV therapeutic intervention strategies and investigations into viral and host factors necessary for virus entry and cell-to-cell spread.


Assuntos
Farmacorresistência Viral/genética , Mutação , Vírus Sinciciais Respiratórios/genética , Animais , Antivirais/toxicidade , Chlorocebus aethiops , Farmacorresistência Viral/imunologia , Células Hep G2 , Humanos , Palivizumab/toxicidade , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/imunologia , Vírus Sinciciais Respiratórios/isolamento & purificação , Vírus Sinciciais Respiratórios/patogenicidade , Genética Reversa/métodos , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...