Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.204
Filtrar
1.
Hum Vaccin Immunother ; 20(1): 2404752, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-39400298

RESUMEN

Cigarette smoking confers additional risk from influenza. This study assessed the effect of smoking on humoral immune response to influenza vaccine. Adults ≥50 y of age were enrolled during the 2011-2016 influenza vaccination seasons in an observational prospective study. Non-fasting whole blood samples for hemagglutination inhibition (HAI) assays were obtained from participants at pre- and 28 d post-clinically administered, trivalent influenza vaccination. Among 273 participants, 133 subjects self-reported as never smokers, 87 as ex-smokers, and 53 as current smokers. Postvaccination geometric mean HAI titers were significantly higher among smokers for A/H1N1 (p = .031) and A/H3N2 (p = .001). Relative to never smokers, smoking was independently related to seroconversion to A/H1N1, A/H3N2 and B. The adjusted odd ratios (ORs) were 5.2 [95% confidence interval (CI), 2.3, 11.5] for seroconversion to A/H1N1, 5.4 (95% CI, 2.4, 12.1) for A/H3N2, and 2.7 (95% CI, 1.3, 5.7) for B. Smoking was also independently related to seroprotection to A/H1N1, A/H3N2 and B. The ORs were 3.6 (95% CI, 1.6, 8.08) for seroprotection to A/H1N1 in smokers, 2.7 (95% CI, 1.14, 6.5) for A/H3N2, and 2.5 (95% CI, 1.1, 5.7) for B. Although the mechanism is unclear, smokers showed a better immune response to influenza vaccination than never smokers and ex-smokers. The results can be used to emphasize the value of influenza vaccination for smokers.


Asunto(s)
Anticuerpos Antivirales , Pruebas de Inhibición de Hemaglutinación , Subtipo H1N1 del Virus de la Influenza A , Subtipo H3N2 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Humanos , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Masculino , Persona de Mediana Edad , Femenino , Estudios Prospectivos , Gripe Humana/prevención & control , Gripe Humana/inmunología , Anticuerpos Antivirales/sangre , Subtipo H1N1 del Virus de la Influenza A/inmunología , Anciano , Subtipo H3N2 del Virus de la Influenza A/inmunología , Fumar/inmunología , Virus de la Influenza B/inmunología , Seroconversión , Vacunación , Inmunidad Humoral
2.
Med Sci Monit ; 30: e945002, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39285625

RESUMEN

BACKGROUND The purpose of the study was to determine the level of antihemagglutinin antibodies in the serum of patients in the geriatric population in Doctor's Surgery NZOZ Nucleus Warsaw, Poland, during the epidemic season 2021/2022 using the hemagglutination inhibition assay (HAI), according to anti-influenza and anti-COVID-19 vaccination, age, and sex. MATERIAL AND METHODS Serum samples taken from 256 patients aged 65 to 99 years were examined for anti-hemagglutinin antibodies and protective levels of antibodies against antigens: A/Victoria/2570/2019 (H1N1)pdm09, A/Cambodia/e0826360/2020(H3N2), B/Washington/02/2019 (B/Victoria lineage), and B/Phuket/3073/2013 (B/Yamagata lineage) of the quadrivalent influenza vaccine for epidemic season 2021/2022. RESULTS The highest protective level, ie, the percentage of people with antibody titers ≥40 was 87.5% and was recorded for subtype A/Cambodia/e0826360/2020(H3N2), the dominant type causing infections in the epidemic season 2021/2022 confirmed by molecular biology methods. Geometric mean titer (GMT) values and protective levels for B/Washington/02/2019 (B/Victoria lineage) antigen were higher for men than women (respectively 38.4 vs 67.6; P<0.001 and 58.0% vs 74.6%; P<0.001). The protective levels of antibodies among patients vaccinated vs unvaccinated against COVID-19 were higher for B/Washington/02/2019 (B/Victoria lineage) and B/Phuket/3073/2013 (B/Yamagata lineage) antigens (64.2% vs 44.4%; P=0.023 and 78.6% vs 55.6%; P=0.004). GMT values for vaccinated against COVID-19 were also higher. There were no significant differences between younger (65-79 years) and older (≥80 years) seniors. CONCLUSIONS The analysis shows differences in the level of individual antibodies, GMT and the protective level depending on subtypes of influenza A or B virus, B/Victoria or B/Yamagata lineage, sex, and previous vaccination history against influenza and COVID-19.


Asunto(s)
Anticuerpos Antivirales , COVID-19 , Vacunas contra la Influenza , Gripe Humana , SARS-CoV-2 , Humanos , Anciano , Polonia/epidemiología , Masculino , Femenino , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Gripe Humana/prevención & control , Gripe Humana/inmunología , Gripe Humana/epidemiología , Vacunas contra la Influenza/inmunología , Anciano de 80 o más Años , COVID-19/prevención & control , COVID-19/inmunología , COVID-19/epidemiología , SARS-CoV-2/inmunología , Vacunas contra la COVID-19/inmunología , Factores Sexuales , Vacunación , Subtipo H3N2 del Virus de la Influenza A/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Factores de Edad , Subtipo H1N1 del Virus de la Influenza A/inmunología , Pruebas de Inhibición de Hemaglutinación/métodos , Estaciones del Año
3.
Cell Genom ; 4(9): 100636, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39197446

RESUMEN

Asthma is a complex disease caused by genetic and environmental factors. Studies show that wheezing during rhinovirus infection correlates with childhood asthma development. Over 150 non-coding risk variants for asthma have been identified, many affecting gene regulation in T cells, but the effects of most risk variants remain unknown. We hypothesized that airway epithelial cells could also mediate genetic susceptibility to asthma given they are the first line of defense against respiratory viruses and allergens. We integrated genetic data with transcriptomics of airway epithelial cells subject to different stimuli. We demonstrate that rhinovirus infection significantly upregulates childhood-onset asthma-associated genes, particularly in non-ciliated cells. This enrichment is also observed with influenza infection but not with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or cytokine activation. Overall, our results suggest that rhinovirus infection is an environmental factor that interacts with genetic risk factors through non-ciliated airway epithelial cells to drive childhood-onset asthma.


Asunto(s)
Asma , Células Epiteliales , Predisposición Genética a la Enfermedad , Infecciones por Picornaviridae , Rhinovirus , Humanos , Asma/genética , Asma/virología , Asma/inmunología , Células Epiteliales/virología , Células Epiteliales/metabolismo , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/virología , Niño , Factores de Riesgo , SARS-CoV-2 , Gripe Humana/genética , Gripe Humana/inmunología , Gripe Humana/virología , COVID-19/genética , COVID-19/virología , COVID-19/inmunología
4.
Clin Infect Dis ; 79(4): 1088-1098, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39099085

RESUMEN

BACKGROUND: We evaluated co-administration of adjuvanted seasonal quadrivalent influenza vaccine (FLU-aQIV) and respiratory syncytial virus (RSV) prefusion F protein-based vaccine (RSVPreF3 OA) in ≥65-year-olds. METHODS: This phase 3, open-label trial randomized ≥65-year-olds to receive FLU-aQIV and RSVPreF3 OA concomitantly (Co-Ad) or sequentially, 1 month apart (Control). Primary objectives were to demonstrate the non-inferiority of FLU-aQIV and RSVPreF3 OA co-administration versus sequential administration in terms of hemagglutination inhibition (HI) titers for each FLU-aQIV strain and RSV-A and RSV-B neutralization titers, 1 month post-vaccination. Reactogenicity and safety were also assessed. RESULTS: Overall, 1045 participants were vaccinated (Co-Ad: 523; Control: 522). Non-inferiority of FLU-aQIV and RSVPreF3 OA co-administration versus sequential administration was demonstrated in terms of HI titers for the A/Victoria(H1N1), B/Victoria, and B/Yamagata influenza strains and RSV-A neutralization titers (upper limits [ULs] of 95% confidence intervals [CIs] for adjusted geometric mean titer [GMT] ratios [Control/Co-Ad] ≤1.50) but not for A/Darwin(H3N2) HI titers (95% CI UL = 1.53). The immune response to A/Darwin(H3N2) was further assessed post-hoc using a microneutralization assay; the post-vaccination adjusted GMT ratio (Control/Co-Ad) was 1.23 (95% CI: 1.06-1.42, ie, UL ≤1.50), suggesting an adequate immune response to A/Darwin(H3N2) following co-administration. RSV-B neutralization titers were comparable between groups (95% CI UL for adjusted GMT ratio ≤1.50). Solicited adverse events were mostly mild or moderate and transient; unsolicited and serious adverse event rates were balanced between groups. CONCLUSIONS: Adjuvanted FLU-aQIV and RSVPreF3 OA had acceptable reactogenicity/safety profiles when co-administered in ≥65-year-olds, without clinically relevant interference with the immune responses to either vaccine. CLINICAL TRIALS REGISTRATION: NCT05568797.


Asunto(s)
Anticuerpos Antivirales , Vacunas contra la Influenza , Gripe Humana , Infecciones por Virus Sincitial Respiratorio , Vacunas contra Virus Sincitial Respiratorio , Humanos , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/efectos adversos , Vacunas contra la Influenza/administración & dosificación , Femenino , Masculino , Anciano , Anticuerpos Antivirales/sangre , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Gripe Humana/prevención & control , Gripe Humana/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/efectos adversos , Virus Sincitial Respiratorio Humano/inmunología , Inmunogenicidad Vacunal , Pruebas de Inhibición de Hemaglutinación , Anciano de 80 o más Años , Proteínas Virales de Fusión/inmunología , Proteínas Virales de Fusión/administración & dosificación , Anticuerpos Neutralizantes/sangre , Vacunación/métodos , Virus de la Influenza B/inmunología
5.
Clin Epigenetics ; 16(1): 114, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39169387

RESUMEN

BACKGROUND: The effect of vaccination on the epigenome remains poorly characterized. In previous research, we identified an association between seroprotection against influenza and DNA methylation at sites associated with the RIG-1 signaling pathway, which recognizes viral double-stranded RNA and leads to a type I interferon response. However, these studies did not fully account for confounding factors including age, gender, and BMI, along with changes in cell-type composition. RESULTS: Here, we studied the influenza vaccine response in a longitudinal cohort vaccinated over two consecutive years (2019-2020 and 2020-2021), using peripheral blood mononuclear cells and a targeted DNA methylation approach. To address the effects of multiple factors on the epigenome, we designed a multivariate multiple regression model that included seroprotection levels as quantified by the hemagglutination-inhibition (HAI) assay test. CONCLUSIONS: Our findings indicate that 179 methylation sites can be combined as potential signatures to predict seroprotection. These sites were not only enriched for genes involved in the regulation of the RIG-I signaling pathway, as found previously, but also enriched for other genes associated with innate immunity to viruses and the transcription factor binding sites of BRD4, which is known to impact T cell memory. We propose a model to suggest that the RIG-I pathway and BRD4 could potentially be modulated to improve immunization strategies.


Asunto(s)
Metilación de ADN , Inmunidad Innata , Vacunas contra la Influenza , Gripe Humana , Humanos , Metilación de ADN/genética , Metilación de ADN/efectos de los fármacos , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Inmunidad Innata/genética , Femenino , Masculino , Gripe Humana/prevención & control , Gripe Humana/inmunología , Gripe Humana/genética , Persona de Mediana Edad , Adulto , Transducción de Señal , Linfocitos T/inmunología , Estudios Longitudinales , Epigénesis Genética , Vacunación , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo
6.
Front Immunol ; 15: 1443096, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39176097

RESUMEN

Introduction: Influenza virus infection can cause a range of clinical symptoms, including respiratory failure (RF) and even death. The mechanisms responsible for the most severe forms of the disease are not yet well understood. The objective is to assess the initial immune response upon admission and its potential impact on infection progression. Methods: We conducted a prospective observational study of patients with influenza virus infection who required admission to a tertiary hospital in the 2017/18 and 2018/19 flu seasons. Immune markers, surrogate markers of neutrophil activation, and blood levels of DNase I and Apolipoprotein-H (ApoH) were determined in the first serum sample available during hospital care. Patients were followed until hospital discharge or death. Initially, 792 patients were included. From this group, 107 patients with poor evolution were selected, and a random control group was matched by day of admission. Results: Patients with poor outcomes had significantly reduced ApoH levels, a soluble protein that regulate both complement and coagulation pathways. In multivariate analysis, low plasma levels of ApoH (OR:5.43; 2.21-13.4), high levels of C- reactive protein (OR:2.73: 1.28-5.4), hyperferritinemia (OR:2.83; 1.28-5.4) and smoking (OR:3.41; 1.04-11.16), were significantly associated with a worse prognosis. RF was independently associated with low levels of ApoH (OR: 5.12; 2.02-1.94), while high levels of IL15 behaved as a protective factor (OR:0.30; 0.12-0.71). Discussion: Therefore, in hospitalized influenza patients, a dysregulated early immune response is associated with a worse outcome. Adequate plasma levels of ApoH are protective against severe influenza and RF and High levels of IL15 protect against RF.


Asunto(s)
Biomarcadores , Gripe Humana , Interleucina-15 , Interleucina-8 , Humanos , Gripe Humana/inmunología , Gripe Humana/sangre , Masculino , Femenino , Biomarcadores/sangre , Pronóstico , Persona de Mediana Edad , Interleucina-15/sangre , Anciano , Estudios Prospectivos , Interleucina-8/sangre , Adulto
7.
Cell Mol Life Sci ; 81(1): 355, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39158695

RESUMEN

Caspase-8, an aspartate-specific cysteine protease that primarily functions as an initiator caspase to induce apoptosis, can downregulate innate immunity in part by cleaving RIPK1 and IRF3. However, patients with caspase-8 mutations or deficiency develop immunodeficiency and are prone to viral infections. The molecular mechanism underlying this controversy remains unknown. Whether caspase-8 enhances or suppresses antiviral responses against influenza A virus (IAV) infection remains to be determined. Here, we report that caspase-8 is readily activated in A549 and NL20 cells infected with the H5N1, H5N6, and H1N1 subtypes of IAV. Surprisingly, caspase-8 deficiency and two caspase-8 inhibitors, Z-VAD and Z-IETD, do not enhance but rather downregulate antiviral innate immunity, as evidenced by decreased TBK1, IRF3, IκBα, and p65 phosphorylation, decreased IL-6, IFN-ß, MX1, and ISG15 gene expression; and decreased IFN-ß production but increased virus replication. Mechanistically, caspase-8 cleaves and inactivates CYLD, a tumor suppressor that functions as a deubiquitinase. Caspase-8 inhibition suppresses CYLD cleavage, RIG-I and TAK1 ubiquitination, and innate immune signaling. In contrast, CYLD deficiency enhances IAV-induced RIG-I and TAK1 ubiquitination and innate antiviral immunity. Neither caspase-3 deficiency nor treatment with its inhibitor Z-DEVD affects CYLD cleavage or antiviral innate immunity. Our study provides evidence that caspase-8 activation in two human airway epithelial cell lines does not silence but rather enhances innate immunity by inactivating CYLD.


Asunto(s)
Caspasa 8 , Proteína 58 DEAD Box , Enzima Desubiquitinante CYLD , Inmunidad Innata , Virus de la Influenza A , Gripe Humana , Quinasas Quinasa Quinasa PAM , Ubiquitinación , Humanos , Enzima Desubiquitinante CYLD/metabolismo , Enzima Desubiquitinante CYLD/genética , Caspasa 8/metabolismo , Caspasa 8/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/inmunología , Virus de la Influenza A/inmunología , Proteína 58 DEAD Box/metabolismo , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/inmunología , Gripe Humana/inmunología , Gripe Humana/virología , Células A549 , Animales , Transducción de Señal/inmunología , Receptores Inmunológicos
8.
J Virol ; 98(8): e0078124, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39078191

RESUMEN

Influenza remains a worldwide public health threat. Although seasonal influenza vaccines are currently the best means of preventing severe disease, the standard-of-care vaccines require frequent updating due to antigenic drift and can have low efficacy, particularly in vulnerable populations. Here, we demonstrate that a single administration of a recombinant adenovirus-associated virus (rAAV) vector expressing a computationally optimized broadly reactive antigen (COBRA)-derived influenza H1 hemagglutinin (HA) induces strongly neutralizing and broadly protective antibodies in naïve mice and ferrets with pre-existing influenza immunity. Following a lethal viral challenge, the rAAV-COBRA vaccine allowed for significantly reduced viral loads in the upper and lower respiratory tracts and complete protection from morbidity and mortality that lasted for at least 5 months post-vaccination. We observed no signs of antibody waning during this study. CpG motif enrichment of the antigen can act as an internal adjuvant to further enhance the immune responses to allow for lower vaccine dosages with the induction of unique interferon-producing CD4+ and CD8+ T cells specific to HA head and stem peptide sequences. Our studies highlight the utility of rAAV as an effective platform to improve seasonal influenza vaccines. IMPORTANCE: Developing an improved seasonal influenza vaccine remains an ambitious goal of researchers and clinicians alike. With influenza routinely causing severe epidemics with the potential to rise to pandemic levels, it is critical to create an effective, broadly protective, and durable vaccine to improve public health worldwide. As a potential solution, we created a rAAV viral vector expressing a COBRA-optimized influenza hemagglutinin antigen with modestly enriched CpG motifs to evoke a robust and long-lasting immune response after a single intramuscular dose without needing boosts or adjuvants. Importantly, the rAAV vaccine boosted antibody breadth to future strains in ferrets with pre-existing influenza immunity. Together, our data support further investigation into the utility of viral vectors as a potential avenue to improve our seasonal influenza vaccines.


Asunto(s)
Inmunidad Adaptativa , Anticuerpos Antivirales , Dependovirus , Hurones , Glicoproteínas Hemaglutininas del Virus de la Influenza , Vacunas contra la Influenza , Infecciones por Orthomyxoviridae , Animales , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Ratones , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/inmunología , Dependovirus/genética , Dependovirus/inmunología , Anticuerpos Neutralizantes/inmunología , Humanos , Femenino , Vectores Genéticos , Ratones Endogámicos BALB C , Vacunación , Gripe Humana/prevención & control , Gripe Humana/inmunología , Linfocitos T CD8-positivos/inmunología
9.
Eur J Immunol ; 54(10): e2451045, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39031535

RESUMEN

Efficient identification of human monoclonal antibodies targeting specific antigenic sites is pivotal for advancing vaccines and immunotherapies against infectious diseases and cancer. Existing screening techniques, however, limit our ability to discover monoclonal antibodies with desired specificity. In this study, we introduce a novel method, blocking of binding (BoB) enzyme-linked immunoassay (ELISA), enabling the detection of high-avidity human antibodies directed to defined epitopes. Leveraging BoB-ELISA, we analyzed the antibody response to known epitopes of influenza A hemagglutinin (HA) in the serum of vaccinated donors. Our findings revealed that serum antibodies targeting head epitopes were immunodominant, whereas antibodies against the stem epitope, although subdominant, were highly prevalent. Extending our analysis across multiple HA strains, we examined the cross-reactive antibody response targeting the stem epitope. Importantly, employing BoB-ELISA we identified donors harboring potent heterosubtypic antibodies targeting the HA stem. B-cell clonal analysis of these donors revealed three novel, genealogically independent monoclonal antibodies with broad cross-reactivity to multiple HAs. In summary, we demonstrated that BoB-ELISA is a sensitive technique for measuring B-cell epitope immunogenicity, enabling the identification of novel monoclonal antibodies with implications for enhanced vaccine development and immunotherapies.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Antivirales , Reacciones Cruzadas , Ensayo de Inmunoadsorción Enzimática , Glicoproteínas Hemaglutininas del Virus de la Influenza , Gripe Humana , Humanos , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Anticuerpos Monoclonales/inmunología , Reacciones Cruzadas/inmunología , Anticuerpos Antivirales/inmunología , Gripe Humana/inmunología , Epítopos/inmunología , Vacunas contra la Influenza/inmunología , Virus de la Influenza A/inmunología
10.
Immunity ; 57(8): 1955-1974.e8, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-38964332

RESUMEN

The nasal mucosa is often the initial site of respiratory viral infection, replication, and transmission. Understanding how infection shapes tissue-scale primary and memory responses is critical for designing mucosal therapeutics and vaccines. We generated a single-cell RNA-sequencing atlas of the murine nasal mucosa, sampling three regions during primary influenza infection and rechallenge. Compositional analysis revealed restricted infection to the respiratory mucosa with stepwise changes in immune and epithelial cell subsets and states. We identified and characterized a rare subset of Krt13+ nasal immune-interacting floor epithelial (KNIIFE) cells, which concurrently increased with tissue-resident memory T (TRM)-like cells. Proportionality analysis, cell-cell communication inference, and microscopy underscored the CXCL16-CXCR6 axis between KNIIFE and TRM cells. Secondary influenza challenge induced accelerated and coordinated myeloid and lymphoid responses without epithelial proliferation. Together, this atlas serves as a reference for viral infection in the upper respiratory tract and highlights the efficacy of local coordinated memory responses.


Asunto(s)
Memoria Inmunológica , Células T de Memoria , Mucosa Nasal , Infecciones por Orthomyxoviridae , Animales , Memoria Inmunológica/inmunología , Ratones , Mucosa Nasal/virología , Mucosa Nasal/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Células T de Memoria/inmunología , Células Epiteliales/inmunología , Células Epiteliales/virología , Ratones Endogámicos C57BL , Humanos , Análisis de la Célula Individual , Gripe Humana/inmunología , Gripe Humana/virología , Femenino , Receptores CXCR6/metabolismo , Receptores CXCR6/inmunología , Virus de la Influenza A/inmunología , Virus de la Influenza A/fisiología
11.
Viruses ; 16(6)2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38932148

RESUMEN

The devastating effects of COVID-19 have highlighted the importance of prophylactic and therapeutic strategies to combat respiratory diseases. Stimulator of interferon gene (STING) is an essential component of the host defense mechanisms against respiratory viral infections. Although the role of the cGAS/STING signaling axis in the innate immune response to DNA viruses has been thoroughly characterized, mounting evidence shows that it also plays a key role in the prevention of RNA virus infections. In this study, we investigated the role of STING activation during Influenza virus (IFV) infection. In both mouse bone marrow-derived macrophages and monocytic cell line THP-1 differentiated with PMA, we found that dimeric amidobenzimidazole (diABZI), a STING agonist, had substantial anti-IFV activity against multiple strains of IFV, including A/H1N1, A/H3N2, B/Yamagata, and B/Victoria. On the other hand, a pharmacological antagonist of STING (H-151) or the loss of STING in human macrophages leads to enhanced viral replication but suppressed IFN expression. Furthermore, diABZI was antiviral against IFV in primary air-liquid interface cultures of nasal epithelial cells. Our data suggest that STING agonists may serve as promising therapeutic antiviral agents to combat IFV.


Asunto(s)
Antivirales , Inmunidad Innata , Macrófagos , Proteínas de la Membrana , Animales , Humanos , Inmunidad Innata/efectos de los fármacos , Ratones , Antivirales/farmacología , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/virología , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Células THP-1 , Replicación Viral/efectos de los fármacos , Gripe Humana/inmunología , Gripe Humana/virología , Gripe Humana/tratamiento farmacológico , Perros , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/virología , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/inmunología , Orthomyxoviridae/fisiología , Bencimidazoles/farmacología , Transducción de Señal/efectos de los fármacos
12.
Int J Mol Sci ; 25(12)2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38928290

RESUMEN

Influenza virus infection poses a great threat to human health globally each year. Non-coding RNAs (ncRNAs) in the human genome have been reported to participate in the replication process of the influenza virus, among which there are still many unknowns about Long Intergenic Non-Coding RNAs (LincRNAs) in the cell cycle of viral infections. Here, we observed an increased expression of Linc01615 in A549 cells upon influenza virus PR8 infection, accompanied by the successful activation of the intracellular immune system. The knockdown of Linc01615 using the shRNAs promoted the proliferation of the influenza A virus, and the intracellular immune system was inhibited, in which the expressions of IFN-ß, IL-28A, IL-29, ISG-15, MX1, and MX2 were decreased. Predictions from the catRAPID website suggested a potential interaction between Linc01615 and DHX9. Also, knocking down Linc01615 promoted influenza virus proliferation. The subsequent transcriptome sequencing results indicated a decrease in Linc01615 expression after influenza virus infection when DHX9 was knocked down. Further analysis through cross-linking immunoprecipitation and high-throughput sequencing (CLIP-seq) in HEK293 cells stably expressing DHX9 confirmed the interaction between DHX9 and Linc01615. We speculate that DHX9 may interact with Linc01615 to partake in influenza virus replication and that Linc01615 helps to activate the intracellular immune system. These findings suggest a deeper connection between DHX9 and Linc01615, which highlights the significant role of Linc01615 in the influenza virus replication process. This research provides valuable insights into understanding influenza virus replication and offers new targets for preventing influenza virus infections.


Asunto(s)
ARN Helicasas DEAD-box , Gripe Humana , ARN Largo no Codificante , Replicación Viral , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Células A549 , Células HEK293 , Gripe Humana/virología , Gripe Humana/genética , Gripe Humana/inmunología , Gripe Humana/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Virus de la Influenza A/fisiología , Animales , Perros , Técnicas de Silenciamiento del Gen , Proteínas de Neoplasias
13.
Int J Infect Dis ; 146: 107108, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38795841

RESUMEN

OBJECTIVES: Daratumumab-treated myeloma patients may face increased seasonal influenza risk due to weakened postvaccination immune responses, especially with daratumumab treatment. We aimed to assess humoral responses to boosted influenza vaccination in daratumumab-treated or -untreated patients. METHODS: In a single-center study, we evaluated humoral responses (hemagglutination-inhibition assay) one month following a two-injection (4-weeks apart) influenza vaccination (standard dose) in 84 patients with multiple myeloma (40 with daratumumab in the past year). RESULTS: Seroprotection rates (titer ≥1/40) after the second vaccine injection were low across vaccinal subtypes (except for A-H3N2): 71.3% (A-H3N2), 19.7% (A-H1N1pdm09), 9.9% (B-Victoria), 11.3% (B-Yamagata). Only A-H3N2 seroprotection rates significantly increased with the booster in daratumumab-treated patients (30% (12/40) after one injection vs 55% (22/40) after the boost; P = 0.01).After propensity score weighting, daratumumab was not significantly associated with a reduced likelihood of seroprotection against at least one vaccine strain (OR 0.65 [95% CI: 0.22-1.88]). CONCLUSION: While daratumumab treatment did not lead to a significant reduction in seroprotection rates following influenza vaccination, a booster vaccine injection demonstrated potential benefit for specific strains (A-H3N2) in patients undergoing daratumumab treatment. Nevertheless, the overall low response rates in patients with multiple myeloma necessitates the development of alternative vaccination and prophylaxis strategies.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Antivirales , Subtipo H3N2 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Mieloma Múltiple , Humanos , Mieloma Múltiple/inmunología , Mieloma Múltiple/tratamiento farmacológico , Masculino , Femenino , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Anciano , Persona de Mediana Edad , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales/administración & dosificación , Gripe Humana/prevención & control , Gripe Humana/inmunología , Anticuerpos Antivirales/sangre , Subtipo H3N2 del Virus de la Influenza A/inmunología , Adulto , Anciano de 80 o más Años , Pruebas de Inhibición de Hemaglutinación , Vacunación , Inmunización Secundaria , Subtipo H1N1 del Virus de la Influenza A/inmunología
14.
Lancet Microbe ; 5(7): 645-654, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38729196

RESUMEN

BACKGROUND: Protection afforded by inactivated influenza vaccines can theoretically be improved by inducing T-cell responses to conserved internal influenza A antigens. We assessed whether, in an influenza controlled human infection challenge, susceptible individuals receiving a vaccine boosting T-cell responses would exhibit lower viral load and decreased symptoms compared with placebo recipients. METHODS: In this single centre, randomised, double-blind phase 2 study, healthy adult (aged 18-55 years) volunteers with microneutralisation titres of less than 20 to the influenza A(H3N2) challenge strain were enrolled at an SGS quarantine facility in Antwerp, Belgium. Participants were randomly assigned double-blind using a permuted-block list with a 3:2 allocation ratio to receive 0·5 mL intramuscular injections of modified vaccinia Ankara (MVA) expressing H3N2 nucleoprotein (NP) and matrix protein 1 (M1) at 1·5 × 108 plaque forming units (4·3 × 108 50% tissue culture infectious dose [TCID50]; MVA-NP+M1 group) or saline placebo (placebo group). At least 6 weeks later, participants were challenged intranasally with 0·5 mL of a 1 × 106 TCID50/mL dose of influenza A/Belgium/4217/2015 (H3N2). Nasal swabs were collected twice daily from day 2 until day 11 for viral PCR, and symptoms of influenza were recorded from day 2 until day 11. The primary outcome was to determine the efficacy of MVA-NP+M1 vaccine to reduce the degree of nasopharyngeal viral shedding as measured by the cumulative viral area under the curve using a log-transformed quantitative PCR. This study is registered with ClinicalTrials.gov, NCT03883113. FINDINGS: Between May 2 and Oct 24, 2019, 145 volunteers were enrolled and randomly assigned to the MVA-NP+M1 group (n=87) or the placebo group (n=58). Of these, 118 volunteers entered the challenge period (71 in the MVA-NP+M1 group and 47 in the placebo group) and 117 participants completed the study (71 in the MVA-NP+M1 group and 46 in the placebo group). 78 (54%) of the 145 volunteers were female and 67 (46%) were male. The primary outcome, overall viral load as determined by quantitative PCR, did not show a statistically significant difference between the MVA-NP+M1 (mean 649·7 [95% CI 552·7-746·7) and placebo groups (mean 726·1 [604·0-848·2]; p=0·17). All reported treatment emergent adverse events (TEAEs; 11 in the vaccination phase and 51 in the challenge phase) were grade 1 and 2, except for two grade 3 TEAEs in the placebo group in the challenge phase. A grade 4 second trimester fetal death, considered possibly related to the MVA-NP+M1 vaccination, and an acute psychosis reported in a placebo participant during the challenge phase were reported. INTERPRETATION: The use of an MVA vaccine to expand CD4+ or CD8+ T cells to conserved influenza A antigens in peripheral blood did not affect nasopharyngeal viral load in an influenza H3N2 challenge model in seronegative, healthy adults. FUNDING: Department of Health and Human Services; Administration for Strategic Preparedness and Response; Biomedical Advanced Research and Development Authority; and Barinthus Biotherapeutics.


Asunto(s)
Linfocitos T CD8-positivos , Subtipo H3N2 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Carga Viral , Humanos , Adulto , Bélgica/epidemiología , Método Doble Ciego , Subtipo H3N2 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Femenino , Masculino , Adulto Joven , Persona de Mediana Edad , Linfocitos T CD8-positivos/inmunología , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Adolescente , Proteínas de la Matriz Viral/inmunología , Proteínas del Núcleo Viral/inmunología , Proteínas de Unión al ARN/inmunología , Proteínas de Unión al ARN/genética , Vacunas de ADN/inmunología , Vacunas de ADN/administración & dosificación , Proteínas de la Nucleocápside/inmunología , Anticuerpos Antivirales/sangre , Inmunidad Celular
15.
Nat Commun ; 15(1): 3666, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38693120

RESUMEN

Respiratory viral infection increases host susceptibility to secondary bacterial infections, yet the precise dynamics within airway epithelia remain elusive. Here, we elucidate the pivotal role of CD47 in the airway epithelium during bacterial super-infection. We demonstrated that upon influenza virus infection, CD47 expression was upregulated and localized on the apical surface of ciliated cells within primary human nasal or bronchial epithelial cells. This induced CD47 exposure provided attachment sites for Staphylococcus aureus, thereby compromising the epithelial barrier integrity. Through bacterial adhesion assays and in vitro pull-down assays, we identified fibronectin-binding proteins (FnBP) of S. aureus as a key component that binds to CD47. Furthermore, we found that ciliated cell-specific CD47 deficiency or neutralizing antibody-mediated CD47 inactivation enhanced in vivo survival rates. These findings suggest that interfering with the interaction between airway epithelial CD47 and pathogenic bacterial FnBP holds promise for alleviating the adverse effects of super-infection.


Asunto(s)
Antígeno CD47 , Células Epiteliales , Infecciones Estafilocócicas , Staphylococcus aureus , Sobreinfección , Antígeno CD47/metabolismo , Antígeno CD47/genética , Humanos , Animales , Sobreinfección/microbiología , Ratones , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/virología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Gripe Humana/metabolismo , Gripe Humana/inmunología , Gripe Humana/virología , Adhesión Bacteriana , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/virología , Ratones Endogámicos C57BL , Bronquios/metabolismo , Bronquios/citología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Ratones Noqueados , Subtipo H1N1 del Virus de la Influenza A
16.
Curr Opin Virol ; 66: 101409, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38564993

RESUMEN

Influenza A virus (IAV) infections pose a global health challenge that necessitates a comprehensive understanding of the host immune response to devise effective therapeutic interventions. As monocytes and macrophages play crucial roles in host defence, inflammation, and repair, this review explores the intricate journey of these cells during and after IAV infection. First, we highlight the dynamics and functions of lung-resident macrophage populations post-IAV. Second, we review the current knowledge of recruited monocytes and monocyte-derived cells, emphasising their roles in viral clearance, inflammation, immunomodulation, and tissue repair. Third, we shed light on the consequences of IAV-induced macrophage alterations on long-term lung immunity. We conclude by underscoring current knowledge gaps and exciting prospects for future research in unravelling the complexities of macrophage responses to respiratory viral infections.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Macrófagos , Monocitos , Humanos , Monocitos/inmunología , Monocitos/virología , Gripe Humana/inmunología , Gripe Humana/virología , Virus de la Influenza A/inmunología , Virus de la Influenza A/fisiología , Macrófagos/inmunología , Macrófagos/virología , Animales , Pulmón/virología , Pulmón/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Inflamación/inmunología , Inflamación/virología
17.
J Med Virol ; 96(4): e29605, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38634474

RESUMEN

Interferon lambda (IFNλ), classified as a type III IFN, is a representative cytokine that plays an important role in innate immunity along with type I IFN. IFNλ can elicit antiviral states by inducing peculiar sets of IFN-stimulated genes (ISGs). In this study, an adenoviral vector expression system with a tetracycline operator system was used to express human IFNλ4 in cells and mice. The formation of recombinant adenovirus (rAd-huIFNλ4) was confirmed using immunohistochemistry assays and transmission electron microscopy. Its purity was verified by quantifying host cell DNA and host cell proteins, as well as by confirming the absence of the replication-competent adenovirus. The transduction of rAd-huIFNλ4 induced ISGs and inhibited four subtypes of the influenza virus in both mouse-derived (LA-4) and human-derived cells (A549). The antiviral state was confirmed in BALB/c mice following intranasal inoculation with 109 PFU of rAd-huIFNλ4, which led to the inhibition of four subtypes of the influenza virus in mouse lungs, with reduced inflammatory lesions. These results imply that human IFNλ4 could induce antiviral status by modulating ISG expression in mice.


Asunto(s)
Antivirales , Gripe Humana , Interferón lambda , Orthomyxoviridae , Animales , Humanos , Ratones , Antivirales/farmacología , Inmunidad Innata , Gripe Humana/inmunología , Gripe Humana/prevención & control , Interferón lambda/metabolismo , Interferón lambda/farmacología , Interferón Tipo I/genética , Interferones/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Vectores Genéticos
18.
J Agric Food Chem ; 72(17): 9782-9794, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38597360

RESUMEN

Uncontrolled inflammation contributes significantly to the mortality in acute respiratory infections. Our previous research has demonstrated that maize bran feruloylated oligosaccharides (FOs) possess notable anti-inflammatory properties linked to the NF-kB pathway regulation. In this study, we clarified that the oral administration of FOs moderately inhibited H1N1 virus infection and reduced lung inflammation in influenza-infected mice by decreasing a wide spectrum of cytokines (IFN-α, IFN-ß, IL-6, IL-10, and IL-23) in the lungs. The mechanism involves FOs suppressing the transduction of the RIG-I/MAVS/TRAF3 signaling pathway, subsequently lowering the expression of NF-κB. In silico analysis suggests that FOs have a greater binding affinity for the RIG-I/MAVS signaling complex. This indicates that FOs have potential as promising targets for immune modulation. Moreover, in MAVS knockout mice, we confirmed that the anti-inflammatory function of FOs against influenza depends on MAVS. Comprehensive analysis using 16S rRNA gene sequencing and metabolite profiling techniques showed that FOs have the potential to restore immunity by modulating the gut microbiota. In conclusion, our study demonstrates that FOs are effective anti-inflammatory phytochemicals in inhibiting lung inflammation caused by influenza. This suggests that FOs could serve as a potential nutritional strategy for preventing the H1N1 virus infection and associated lung inflammation.


Asunto(s)
Proteína 58 DEAD Box , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Ratones Noqueados , Oligosacáridos , Infecciones por Orthomyxoviridae , Transducción de Señal , Factor 3 Asociado a Receptor de TNF , Animales , Ratones , Oligosacáridos/administración & dosificación , Oligosacáridos/química , Oligosacáridos/farmacología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/metabolismo , Subtipo H1N1 del Virus de la Influenza A/inmunología , Humanos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Gripe Humana/metabolismo , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor 3 Asociado a Receptor de TNF/inmunología , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Proteína 58 DEAD Box/inmunología , Neumonía/inmunología , Neumonía/prevención & control , Neumonía/metabolismo , Neumonía/virología , Ratones Endogámicos C57BL , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Pulmón/virología , Citocinas/metabolismo , Citocinas/inmunología , Citocinas/genética , Femenino , FN-kappa B/inmunología , FN-kappa B/genética , FN-kappa B/metabolismo , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología
19.
J Biol Chem ; 300(4): 107153, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38462163

RESUMEN

The innate immune system features a web of interacting pathways that require exquisite regulation. To identify novel nodes in this immune landscape, we conducted a gain-of-function, genome-wide CRISPR activation screen with influenza A virus. We identified both appreciated and novel antiviral genes, including Jade family PHD zinc finger 3 (JADE3) a protein involved in directing the histone acetyltransferase histone acetyltransferase binding to ORC1 complex to modify chromatin and regulate transcription. JADE3 is both necessary and sufficient to restrict influenza A virus infection. Our results suggest a distinct function for JADE3 as expression of the closely related paralogs JADE1 and JADE2 does not confer resistance to influenza A virus infection. JADE3 is required for both constitutive and inducible expression of the well-characterized antiviral gene interferon-induced transmembrane protein 3 (IFITM3). Furthermore, we find JADE3 activates the NF-kB signaling pathway, which is required for the promotion of IFITM3 expression by JADE3. Therefore, we propose JADE3 activates an antiviral genetic program involving NF-kB-dependent IFITM3 expression to restrict influenza A virus infection.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad Innata , Proteínas de la Membrana , FN-kappa B , Proteínas Oncogénicas , Proteínas de Unión al ARN , Animales , Humanos , Sistemas CRISPR-Cas , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Células HEK293 , Inmunidad Innata/genética , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Transducción de Señal , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/inmunología
20.
Nature ; 621(7980): 813-820, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37587341

RESUMEN

Disruption of the lung endothelial-epithelial cell barrier following respiratory virus infection causes cell and fluid accumulation in the air spaces and compromises vital gas exchange function1. Endothelial dysfunction can exacerbate tissue damage2,3, yet it is unclear whether the lung endothelium promotes host resistance against viral pathogens. Here we show that the environmental sensor aryl hydrocarbon receptor (AHR) is highly active in lung endothelial cells and protects against influenza-induced lung vascular leakage. Loss of AHR in endothelia exacerbates lung damage and promotes the infiltration of red blood cells and leukocytes into alveolar air spaces. Moreover, barrier protection is compromised and host susceptibility to secondary bacterial infections is increased when endothelial AHR is missing. AHR engages tissue-protective transcriptional networks in endothelia, including the vasoactive apelin-APJ peptide system4, to prevent a dysplastic and apoptotic response in airway epithelial cells. Finally, we show that protective AHR signalling in lung endothelial cells is dampened by the infection itself. Maintenance of protective AHR function requires a diet enriched in naturally occurring AHR ligands, which activate disease tolerance pathways in lung endothelia to prevent tissue damage. Our findings demonstrate the importance of endothelial function in lung barrier immunity. We identify a gut-lung axis that affects lung damage following encounters with viral pathogens, linking dietary composition and intake to host fitness and inter-individual variations in disease outcome.


Asunto(s)
Células Endoteliales , Pulmón , Infecciones por Orthomyxoviridae , Receptores de Hidrocarburo de Aril , Animales , Humanos , Ratones , Apelina/metabolismo , Dieta , Células Endoteliales/metabolismo , Endotelio/citología , Endotelio/metabolismo , Células Epiteliales/metabolismo , Eritrocitos/metabolismo , Gripe Humana/inmunología , Gripe Humana/metabolismo , Intestinos/metabolismo , Leucocitos/metabolismo , Ligandos , Pulmón/inmunología , Pulmón/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA