RESUMEN
SARS-CoV-2 JN.1 with an additional L455S mutation on spike when compared with its parental variant BA.2.86 has outcompeted all earlier variants to become the dominant circulating variant. Recent studies investigated the immune resistance of SARS-CoV-2 JN.1 but additional factors are speculated to contribute to its global dominance, which remain elusive until today. Here, we find that SARS-CoV-2 JN.1 has a higher infectivity than BA.2.86 in differentiated primary human nasal epithelial cells (hNECs). Mechanistically, we demonstrate that the gained infectivity of SARS-CoV-2 JN.1 over BA.2.86 associates with increased entry efficiency conferred by L455S and better spike cleavage in hNECs. Structurally, S455 altered the mode of binding of JN.1 spike protein to ACE2 when compared to BA.2.86 spike at ACE2H34, and modified the internal structure of JN.1 spike protein by increasing the number of hydrogen bonds with neighboring residues. These findings indicate that a single mutation (L455S) enhances virus entry in hNECs and increases immune evasiveness, which contribute to the robust transmissibility of SARS-CoV-2 JN.1. We further evaluate the in vitro and in vivo virological characteristics between SARS-CoV-2 BA.2.86/JN.1 and EG.5.1/HK.3, and identify key lineage-specific features of the two Omicron sublineages that contribute to our understanding on Omicron antigenicity, transmissibility, and pathogenicity.
Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Evasión Inmune , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Humanos , SARS-CoV-2/genética , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Evasión Inmune/genética , COVID-19/virología , COVID-19/inmunología , Animales , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Internalización del Virus , Mutación , Ratones , Mucosa Nasal/virología , Mucosa Nasal/inmunología , Células Epiteliales/virología , Células Epiteliales/inmunología , Chlorocebus aethiops , Femenino , Células VeroRESUMEN
Equine herpesvirus type 1 (EHV-1) enters through the upper respiratory tract (URT). Mucosal immunity at the URT is crucial in limiting viral infection and morbidity. Here, intranasal immune cells were collected from horses (n = 15) during an experimental EHV-1 infection. CD4+ and CD8+ T cells were the major intranasal cell populations before infection and increased significantly by day six and fourteen post-infection, respectively. Nasal mucosal T cells were further characterized in healthy horses. Compared to peripheral blood mononuclear cells (PBMC), mucosal CD8+ T-cell percentages were elevated, while CD4+ T-cell percentages were similar. A small population of CD4+CD8+ T cells was also recovered from mucosal samples. Within the URT tissue, CD4+ cells predominantly accumulated in the epithelial layer, while most CD8+ cells resided deeper in the mucosa or the submucosa below the basement membrane. In vitro stimulation of mucosal cells from healthy horses with (n = 5) or without (n = 5) peripheral T-cell immunity against EHV-1 induced IFN-γ production in nasal T cells upon polyclonal stimulation. However, after EHV-1 re-stimulation, mucosal T cells failed to respond with IFN-γ. This work provided the first characterization of mucosal T-cell phenotypes and functions in the URT of healthy horses and during EHV-1 infection.
Asunto(s)
Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Infecciones por Herpesviridae , Herpesvirus Équido 1 , Enfermedades de los Caballos , Inmunidad Mucosa , Mucosa Nasal , Animales , Caballos/inmunología , Herpesvirus Équido 1/inmunología , Mucosa Nasal/virología , Mucosa Nasal/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/virología , Linfocitos T CD8-positivos/inmunología , Enfermedades de los Caballos/inmunología , Enfermedades de los Caballos/virología , Linfocitos T CD4-Positivos/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Linfocitos T/inmunología , FemeninoRESUMEN
Virus-specific nasal resident T cells are important for protection against subsequent infection with a similar virus. Here we examine the phenotypes and functions of SARS-CoV-2-specific T cells in the nasal mucosa of vaccinated individuals with breakthrough infection (BTI) or without infection. Nasal tissues are obtained from participants during sinus surgery. Analysis of activation-induced markers implicates that a considerable proportion of spike (S)-reactive nasal CD8+ T cells express CD103, a tissue-resident marker. MHC-I multimer staining is performed to analyze the ex vivo phenotype and function of SARS-CoV-2 S-specific CD8+ T cells. We detect multimer+CD8+ T cells with tissue-resident phenotypes in nasal tissue samples from vaccinees without infection as well as vaccinees with BTI. Multimer+CD8+ T cells remain present in nasal tissues over one year after the last exposure to S antigen, although the frequency decreases. Upon direct ex vivo stimulation with epitope peptides, nasal multimer+CD8+ T cells-particularly the CD49a+ subset-exhibit immediate effector functions, including IFN-γ production. CITE-seq analysis of S-reactive AIM+CD8+ T cells confirms the enhanced effector function of the CD49a+ subset. These findings indicate that among individuals previously exposed to S antigen by vaccination or BTI, S-specific nasal-resident CD49a+CD8+ memory T cells can rapidly respond to SARS-CoV-2 during infection or reinfection.
Asunto(s)
Linfocitos T CD8-positivos , COVID-19 , Interferón gamma , Células T de Memoria , Mucosa Nasal , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Humanos , Glicoproteína de la Espiga del Coronavirus/inmunología , Linfocitos T CD8-positivos/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , SARS-CoV-2/inmunología , Mucosa Nasal/inmunología , Mucosa Nasal/virología , COVID-19/inmunología , COVID-19/virología , Células T de Memoria/inmunología , Masculino , Femenino , Persona de Mediana Edad , Adulto , Integrina alfa1/inmunología , Integrina alfa1/metabolismo , Vacunas contra la COVID-19/inmunología , Antígenos CD/metabolismo , Antígenos CD/inmunología , Memoria Inmunológica/inmunología , Cadenas alfa de IntegrinasRESUMEN
Bovine herpesvirus type 1 (BoHV-1) establishes lifelong latency in trigeminal ganglionic (TG) neurons following intranasal and ocular infection in cattle. Periodically, the latent virus reactivates in the TG due to stress and is transported anterogradely to nerve endings in the nasal epithelium, where the virus replicates and sheds. Consequently, BoHV-1 is transmitted to susceptible animals and maintained in the cattle population. Modified live BoHV-1 vaccine strains (BoHV-1 MLV) also have a similar latency reactivation. Therefore, they circulate and are maintained in cattle herds. Additionally, they can regain virulence and cause vaccine outbreaks because they mutate and recombine with other circulating field wild-type (wt) strains. Recently, we constructed a BoHV-1 quadruple mutant virus (BoHV-1qmv) that lacks immune evasive properties due to UL49.5 and glycoprotein G (gG) deletions. In addition, it also lacks the gE cytoplasmic tail (gE CT) and Us9 gene sequences designed to make it safe, increase its vaccine efficacy against BoHV-1, and restrict its anterograde neuronal transport noted above. Further, we engineered the BoHV-1qmv-vector to serve as a subunit vaccine against the Rift Valley fever virus (BoHV-1qmv Sub-RVFV) (doi: 10.3390/v15112183). In this study, we determined the latency reactivation and nasal virus shedding properties of BoHV-1qmv (vector) and BoHV-1qmv-vectored subunit RVFV (BoHV-1qmv sub-RVFV) vaccine virus in calves in comparison to the BoHV-1 wild-type (wt) following intranasal inoculation. The real-time PCR results showed that BoHV-1 wt- but not the BoHV-1qmv vector- and BoHV-1qmv Sub-RVFV-inoculated calves shed virus in the nose following dexamethasone-induced latency reactivation; however, like the BoHV-1 wt, both the BoHV-1qmv vector and BoHV-1qmv Sub-RVFV viruses established latency, were reactivated, and replicated in the TG neurons. These results are consistent with the anterograde neurotransport function of the gE CT and Us9 sequences, which are deleted in the BoHV-1qmv and BoHV-1qmv Sub-RVFV.
Asunto(s)
Herpesvirus Bovino 1 , Mucosa Nasal , Neuronas , Ganglio del Trigémino , Activación Viral , Latencia del Virus , Esparcimiento de Virus , Animales , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/fisiología , Herpesvirus Bovino 1/inmunología , Bovinos , Mucosa Nasal/virología , Ganglio del Trigémino/virología , Neuronas/virología , Eliminación de Gen , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/genética , Vacunas Atenuadas/administración & dosificación , Replicación Viral , Enfermedades de los Bovinos/virología , Enfermedades de los Bovinos/prevención & control , Enfermedades de los Bovinos/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/genética , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/virología , Infecciones por Herpesviridae/prevención & control , Infecciones por Herpesviridae/inmunología , Vacunas Virales/inmunología , Vacunas Virales/genética , Vectores Genéticos/genética , Rinotraqueítis Infecciosa Bovina/virología , Rinotraqueítis Infecciosa Bovina/prevención & control , Rinotraqueítis Infecciosa Bovina/inmunología , Vacunas contra Herpesvirus/genética , Vacunas contra Herpesvirus/inmunologíaRESUMEN
BACKGROUNDThe level of nasal spike-specific secretory IgA (sIgA) is inversely correlated with the risk of SARS-CoV-2 Omicron infection. This study aimed to evaluate the safety and immunogenicity of intranasal vaccination using Ad5-S-Omicron (NB2155), a replication-incompetent human type 5 adenovirus carrying Omicron BA.1 spike.METHODSAn open-label, single-center, investigator-initiated trial was carried out on 128 health care workers who had never been infected with SARS-CoV-2 and had previously received 2 or 3 injections of inactivated whole-virus vaccines, with the last dose given 3-19 months previously (median 387 days, IQR 333-404 days). Participants received 2 intranasal sprays of NB2155 at 28-day intervals between November 30 and December 30, 2022. Safety was evaluated by solicited adverse events and laboratory tests. The elevation of nasal mucosal spike-specific sIgA and serum neutralizing activities were assessed. All participants were monitored for infection by antigen tests, disease symptoms, and the elevation of nucleocapsid-specific sIgA in the nasal passage.RESULTSThe vaccine-related solicited adverse events were mild. Nasal spike-specific sIgA against 10 strains had a mean geometric mean fold increase of 4.5 after the first dose, but it increased much higher to 51.5 after the second dose. Serum neutralizing titers also increased modestly to 128.1 (95% CI 74.4-220.4) against authentic BA.1 and 76.9 (95% CI 45.4-130.2) against BA.5 at 14 days after the second dose. Due to the lifting of the zero-COVID policy in China on December 7, 2022, 57.3% of participants were infected with BA.5 between days 15 and 28 after the first dose, whereas no participants reported having any symptomatic infections between day 3 and day 90 after the second dose. The elevation of nasal nucleocapsid-specific sIgA on days 0, 14, 42, and 118 after the first dose was assessed to verify that these 2-dose participants had no asymptomatic infections.CONCLUSIONA 2-dose intranasal vaccination regimen using NB2155 was safe, was well tolerated, and could dramatically induce broad-spectrum spike-specific sIgA in the nasal passage. Preliminary data suggested that the intranasal vaccination may establish an effective mucosal immune barrier against infection and warranted further clinical studies.TRIAL REGISTRATIONChinese Clinical Trial Registry (ChiCTR2300070346).FUNDINGNatural Science Foundation of China, Guangzhou Laboratory, The First Affiliated Hospital of Guangzhou Medical University.
Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Inmunidad Mucosa , Inmunoglobulina A Secretora , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adenoviridae , Administración Intranasal , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , COVID-19/prevención & control , COVID-19/inmunología , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/administración & dosificación , Vacunas contra la COVID-19/efectos adversos , Vectores Genéticos/administración & dosificación , Inmunoglobulina A Secretora/inmunología , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunación/métodosRESUMEN
COVID-19 remains a severe condition for many including immunocompromised individuals. There remains a need for effective measures against this and other respiratory infections, which transmit via virus-laden droplets that reach the nasal or oral mucosae. Nasal sprays offer potential protection against viruses. Such formulations should preserve normal nasal mucociliary function. The antiviral barrier efficacy and effects on mucociliary function of astodrimer sodium nasal spray (AS-NS) were evaluated and compared with other available nasal sprays-low pH hydroxypropyl methylcellulose (HPMC-NS), iota-carrageenan (Carr-NS), nitric oxide (NO-NS), and povidone iodine (PI-NS). Assays simulated clinical conditions. Antiviral barrier function and cell viability were assessed in airway cell monolayers, while a model of fully differentiated human nasal epithelium (MucilAir™) was utilized to evaluate tissue integrity, cytotoxicity, cilia beating frequency, and mucociliary clearance. AS-NS reduced infectious virus in cell monolayers and demonstrated a benign cytotoxicity profile. In human nasal epithelium ex vivo, AS-NS had no impact on mucociliary function (cilia beating nor mucociliary clearance). Carr-NS, HPMC-NS, NO-NS and PI-NS demonstrated limited antiviral effects, while HPMC-NS caused inhibition of mucociliary function. Astodrimer sodium nasal spray demonstrates an acceptable nonclinical efficacy and safety profile as a barrier nasal spray against respiratory viral infection in the nasal cavity.
Asunto(s)
Depuración Mucociliar , Mucosa Nasal , Rociadores Nasales , SARS-CoV-2 , Humanos , Mucosa Nasal/virología , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , SARS-CoV-2/efectos de los fármacos , Depuración Mucociliar/efectos de los fármacos , Antivirales/farmacología , Antivirales/administración & dosificación , COVID-19/virología , COVID-19/metabolismo , Tratamiento Farmacológico de COVID-19 , Supervivencia Celular/efectos de los fármacosRESUMEN
This study aimed to investigate the impact of different types of nasal inflammation on the regulation of entry-associated genes of respiratory viruses, including severe acute respiratory syndrome coronavirus 2 (SARS CoV-2), Middle East respiratory syndrome coronavirus (MERS-CoV), human coronavirus 229E (HCoV-229E), and influenza virus, in the nasal epithelium. Subjects were classified into three groups: control, eosinophilic chronic rhinosinusitis (ECRS), and noneosinophilic CRS (NECRS) groups. Angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine subtype 2 (TMPRSS2), alanyl aminopeptidase (ANPEP), dipeptidyl peptidase 4 (DPP4), and beta-galactoside alpha-2,6-sialyltransferase 1 (ST6GAL1), and beta-galactoside alpha-2,3-sialyltransferase 4 (ST3GAL4) were selected as key entry-associated genes for SARS-CoV-2, HCoV-229E, MERS-CoV, and influenza, respectively, and were evaluated. Brushing samples obtained from each group and human nasal epithelial cells cultured using an air-liquid interface system were treated for 7 days with typical inflammatory cytokines and analyzed using real-time polymerase chain reaction. Western blot analysis and confocal microscopy were performed. The entry-associated genes showed distinct regulation patterns in response to each interleukin-4 (IL-4), interleukin-13 (IL-13), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ). Specifically, ACE2 significantly decreased in type 2 cytokines (IL-4 and IL-13), while TMPRSS2 significantly decreased in type 1 cytokines (TNF-α and IFN-γ). ANPEP significantly decreased in both types of cytokines. Remarkably, DPP4 significantly increased in type 2 cytokines and decreased in type 1 cytokines. Moreover, ST6GAL1 and ST3GAL4 significantly increased in type 2 cytokines and decreased in type 1 cytokines, particularly IFN-γ. These findings were supported by western blot analysis and confocal imaging results, especially for ACE2 and DPP4. The findings regarding differential regulation suggest that patients with ECRS, primarily mediated by type 2 inflammation, may have lower susceptibility to SARS-CoV-2 and HCoV-229E infections but higher susceptibility to MERS-CoV and influenza infections.
Asunto(s)
Citocinas , Mucosa Nasal , Internalización del Virus , Humanos , Citocinas/genética , Citocinas/metabolismo , Mucosa Nasal/virología , Adulto , Masculino , Femenino , Persona de Mediana Edad , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Sinusitis/virología , Sinusitis/genética , Sinusitis/inmunología , SARS-CoV-2/inmunología , Rinitis/virología , Rinitis/genética , Rinitis/inmunología , Regulación de la Expresión Génica , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , COVID-19/inmunología , COVID-19/virología , Coronavirus Humano 229E/genética , Dipeptidil Peptidasa 4/genética , Dipeptidil Peptidasa 4/metabolismo , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunologíaRESUMEN
Pregnancy is associated with profound changes in immunity. However, pregnancy-related respiratory immune adaptations in response to influenza infection and their impact on disease severity remain unclear. Here, we describe, in a preclinical model of mid-gestation pregnancy, a mechanism of enhanced host defense against influenza A virus (IAV) localized to the nasal cavity that limits viral replication and reduces the magnitude of intrapulmonary immune responses. Consequently, the pregnant mice show reduced pulmonary pathology and preserved airway function after IAV infection. The early restriction of viral replication is independent of type I interferon (IFN) but dependent on increased antimicrobial peptides (AMPs) driven by interleukin-17+ (IL-17+) γδ+ T cells within the nasal passages. This pathway of host defense against IAV infection in the upper airways during pregnancy restricts early viral infection and prevents virus dissemination into the lung supporting maternal fitness.
Asunto(s)
Virus de la Influenza A , Interferón Tipo I , Interleucina-17 , Mucosa Nasal , Infecciones por Orthomyxoviridae , Animales , Femenino , Embarazo , Interleucina-17/metabolismo , Interleucina-17/inmunología , Ratones , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Mucosa Nasal/metabolismo , Virus de la Influenza A/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Replicación Viral , Pulmón/inmunología , Pulmón/virologíaRESUMEN
Sneezing and coughing are primary symptoms of many respiratory viral infections and allergies. It is generally assumed that sneezing and coughing involve common sensory receptors and molecular neurotransmission mechanisms. Here, we show that the nasal mucosa is innervated by several discrete populations of sensory neurons, but only one population (MrgprC11+MrgprA3-) mediates sneezing responses to a multitude of nasal irritants, allergens, and viruses. Although this population also innervates the trachea, it does not mediate coughing, as revealed by our newly established cough model. Instead, a distinct sensory population (somatostatin [SST+]) mediates coughing but not sneezing, unraveling an unforeseen sensory difference between sneezing and coughing. At the circuit level, sneeze and cough signals are transmitted and modulated by divergent neuropathways. Together, our study reveals the difference in sensory receptors and neurotransmission/modulation mechanisms between sneezing and coughing, offering neuronal drug targets for symptom management in respiratory viral infections and allergies.
Asunto(s)
Tos , Estornudo , Animales , Ratones , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Células Receptoras Sensoriales/virología , Masculino , Mucosa Nasal/virología , Mucosa Nasal/metabolismo , Femenino , Tráquea/virología , Ratones Endogámicos C57BL , Humanos , Receptores Acoplados a Proteínas G/metabolismoRESUMEN
Studies on the immune-regulatory roles played by the commensal microbes residing in the nasal mucosa consider the contribution of antiviral immune responses. Here, we sought to identify the nasal microbiome, Staphylococcus epidermidis-regulated antiviral immune responses and the alteration of polyamine metabolites in nasal epithelium. We found that polyamines were required for the life cycle of influenza A virus (IAV) and depletion of polyamines disturbed IAV replication in normal human nasal epithelial (NHNE) cells. Inoculation of S. epidermidis also suppressed IAV infection and the concentration of polyamines including putrescine, spermidine, and spermine was completely attenuated in S. epidermidis-inoculated NHNE cells. S. epidermidis activated the enzyme involved in the production of ornithine from arginine and downregulated the activity of the enzyme involved in the production of putrescine from ornithine in nasal epithelium. S. epidermidis also induced the activation of enzymes that promote the extracellular export of spermine and spermidine in NHNE cells. Our findings demonstrate that S. epidermidis is shown to be able of creating an intracellular environment lacking polyamines in the nasal epithelium and promote the balance of cellular polyamines in favor of the host to restrict influenza virus replication.
Asunto(s)
Virus de la Influenza A , Mucosa Nasal , Poliaminas , Staphylococcus epidermidis , Simbiosis , Replicación Viral , Staphylococcus epidermidis/fisiología , Staphylococcus epidermidis/metabolismo , Humanos , Poliaminas/metabolismo , Virus de la Influenza A/fisiología , Mucosa Nasal/microbiología , Mucosa Nasal/virología , Mucosa Nasal/metabolismo , Gripe Humana/virología , Gripe Humana/metabolismoRESUMEN
SARS-CoV-2 infection induces the generation of virus-specific CD4+ and CD8+ effector and memory T cells. However, the contribution of T cells in controlling SARS-CoV-2 during infection is not well understood. Following infection of C57BL/6 mice, SARS-CoV-2-specific CD4+ and CD8+ T cells are recruited to the respiratory tract, and a vast proportion secrete the cytotoxic molecule granzyme B. Using depleting antibodies, we found that T cells within the lungs play a minimal role in viral control, and viral clearance occurs in the absence of both CD4+ and CD8+ T cells through 28 days postinfection. In the nasal compartment, depletion of both CD4+ and CD8+ T cells, but not individually, results in persistent, culturable virus replicating in the nasal epithelial layer through 28 days postinfection. Viral sequencing analysis revealed adapted mutations across the SARS-CoV-2 genome, including a large deletion in ORF6. Overall, our findings highlight the importance of T cells in controlling virus replication within the respiratory tract during SARS-CoV-2 infection.
Asunto(s)
Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , COVID-19 , Ratones Endogámicos C57BL , SARS-CoV-2 , Replicación Viral , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , SARS-CoV-2/fisiología , SARS-CoV-2/inmunología , COVID-19/virología , COVID-19/inmunología , COVID-19/prevención & control , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD4-Positivos/metabolismo , Ratones , Pulmón/virología , Pulmón/inmunología , Humanos , Femenino , Mucosa Nasal/virología , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Granzimas/metabolismoRESUMEN
The upper respiratory tract is the initial site of SARS-CoV-2 infection. Nasal spike-specific secretory immunoglobulin A (sIgA) correlates with protection against Omicron breakthrough infection. We report that intranasal vaccination using human adenovirus serotype 5 (Ad5) vectored Omicron spike in people who previously vaccinated with ancestral vaccine could induce robust neutralizing sIgA in the nasal passage. Nasal sIgA was predominantly present in dimeric and multimeric forms and accounted for nearly 40% of total proteins in nasal mucosal lining fluids (NMLFs). A low-level IgG could also be detected in NMLFs but not IgM, IgD, and IgE. After a complete nasal wash, sIgA in the nasal passage could be replenished rapidly within a few hours. A comparison of purified paired serum IgA, serum IgG, and nasal sIgA from the same individuals showed that sIgA was up to 3-logs more potent than serum antibodies in binding to spikes and in neutralizing Omicron subvariants. Serum IgG and IgA failed to neutralize XBB and BA.2.86, while nasal sIgA retained potent neutralization against these newly emerged variants. Further analysis showed that sIgA was more effective than IgG or IgA in blocking spike-mediated cell-to-cell transmission and protecting hACE2 mice from XBB challenge. Using a sIgA monoclonal antibody as a reference, we estimated that the total nasal sIgA contains about 2.6-3.9% spike-specific sIgA in NMLFs collected approximately one month after intranasal vaccination. Our study provided insights for developing intranasal vaccines that can induce sIgA to build an effective and mutation-resistant first-line immune barrier against constantly emerging variants.
Asunto(s)
Administración Intranasal , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Vacunas contra la COVID-19 , COVID-19 , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Humanos , SARS-CoV-2/inmunología , SARS-CoV-2/genética , COVID-19/prevención & control , COVID-19/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Animales , Ratones , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/administración & dosificación , Inmunoglobulina A/inmunología , Inmunoglobulina A/sangre , Inmunoglobulina A/genética , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Femenino , Vectores Genéticos/inmunología , Vectores Genéticos/genética , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , Inmunoglobulina A Secretora/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina G/sangre , MasculinoRESUMEN
BACKGROUND: While inflammatory and immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in peripheral blood are extensively described, responses at the upper respiratory mucosal site of initial infection are relatively poorly defined. We sought to identify mucosal cytokine/chemokine signatures that distinguished coronavirus disease 2019 (COVID-19) severity categories, and relate these to disease progression and peripheral inflammation. METHODS: We measured 35 cytokines and chemokines in nasal samples from 274 patients hospitalized with COVID-19. Analysis considered the timing of sampling during disease, as either the early (0-5 days after symptom onset) or late (6-20 days after symptom onset) phase. RESULTS: Patients that survived severe COVID-19 showed interferon (IFN)-dominated mucosal immune responses (IFN-γ, CXCL10, and CXCL13) early in infection. These early mucosal responses were absent in patients who would progress to fatal disease despite equivalent SARS-CoV-2 viral load. Mucosal inflammation in later disease was dominated by interleukin 2 (IL-2), IL-10, IFN-γ, and IL-12p70, which scaled with severity but did not differentiate patients who would survive or succumb to disease. Cytokines and chemokines in the mucosa showed distinctions from responses evident in the peripheral blood, particularly during fatal disease. CONCLUSIONS: Defective early mucosal antiviral responses anticipate fatal COVID-19 but are not associated with viral load. Early mucosal immune responses may define the trajectory of severe COVID-19.
Asunto(s)
COVID-19 , Citocinas , Inflamación , SARS-CoV-2 , Humanos , COVID-19/inmunología , COVID-19/mortalidad , Masculino , Femenino , Persona de Mediana Edad , SARS-CoV-2/inmunología , Citocinas/sangre , Anciano , Inflamación/inmunología , Inmunidad Mucosa , Carga Viral , Adulto , Quimiocinas/sangre , Índice de Severidad de la Enfermedad , Mucosa Nasal/inmunología , Mucosa Nasal/virologíaRESUMEN
Children resist COVID-19, and previous studies reported increased innate immunity in their upper airways. A new paper by Watkins et al. (https://doi.org/10.1084/jem.20230911) shows that the nasal mucosa of children is characterized by often asymptomatic viral and/or bacterial infections that dynamically regulate distinct innate immune programs.
Asunto(s)
COVID-19 , Inmunidad Innata , Mucosa Nasal , SARS-CoV-2 , Humanos , COVID-19/inmunología , Niño , Inmunidad Innata/inmunología , SARS-CoV-2/inmunología , Mucosa Nasal/virología , Mucosa Nasal/inmunologíaRESUMEN
Studies during the COVID-19 pandemic showed that children had heightened nasal innate immune responses compared with adults. To evaluate the role of nasal viruses and bacteria in driving these responses, we performed cytokine profiling and comprehensive, symptom-agnostic testing for respiratory viruses and bacterial pathobionts in nasopharyngeal samples from children tested for SARS-CoV-2 in 2021-22 (n = 467). Respiratory viruses and/or pathobionts were highly prevalent (82% of symptomatic and 30% asymptomatic children; 90 and 49% for children <5 years). Virus detection and load correlated with the nasal interferon response biomarker CXCL10, and the previously reported discrepancy between SARS-CoV-2 viral load and nasal interferon response was explained by viral coinfections. Bacterial pathobionts correlated with a distinct proinflammatory response with elevated IL-1ß and TNF but not CXCL10. Furthermore, paired samples from healthy 1-year-olds collected 1-2 wk apart revealed frequent respiratory virus acquisition or clearance, with mucosal immunophenotype changing in parallel. These findings reveal that frequent, dynamic host-pathogen interactions drive nasal innate immune activation in children.
Asunto(s)
COVID-19 , Inmunidad Innata , SARS-CoV-2 , Humanos , Inmunidad Innata/inmunología , Preescolar , Lactante , COVID-19/inmunología , COVID-19/virología , Niño , SARS-CoV-2/inmunología , Femenino , Masculino , Nasofaringe/inmunología , Nasofaringe/virología , Nasofaringe/microbiología , Carga Viral , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Mucosa Nasal/microbiología , Citocinas/metabolismo , Citocinas/inmunología , Interacciones Huésped-Patógeno/inmunología , Adolescente , Nariz/inmunología , Nariz/virología , Nariz/microbiología , Coinfección/inmunología , Coinfección/virologíaRESUMEN
The upper airway is an important site of infection, but immune memory in the human upper airway is poorly understood, with implications for COVID-19 and many other human diseases1-4. Here we demonstrate that nasal and nasopharyngeal swabs can be used to obtain insights into these challenging problems, and define distinct immune cell populations, including antigen-specific memory B cells and T cells, in two adjacent anatomical sites in the upper airway. Upper airway immune cell populations seemed stable over time in healthy adults undergoing monthly swabs for more than 1 year, and prominent tissue resident memory T (TRM) cell and B (BRM) cell populations were defined. Unexpectedly, germinal centre cells were identified consistently in many nasopharyngeal swabs. In subjects with SARS-CoV-2 breakthrough infections, local virus-specific BRM cells, plasma cells and germinal centre B cells were identified, with evidence of local priming and an enrichment of IgA+ memory B cells in upper airway compartments compared with blood. Local plasma cell populations were identified with transcriptional profiles of longevity. Local virus-specific memory CD4+ TRM cells and CD8+ TRM cells were identified, with diverse additional virus-specific T cells. Age-dependent upper airway immunological shifts were observed. These findings provide new understanding of immune memory at a principal mucosal barrier tissue in humans.
Asunto(s)
Memoria Inmunológica , Células B de Memoria , Células T de Memoria , Mucosa Nasal , Nasofaringe , SARS-CoV-2 , Adulto , Humanos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/citología , COVID-19/inmunología , COVID-19/virología , Centro Germinal/inmunología , Centro Germinal/citología , Inmunoglobulina A/inmunología , Memoria Inmunológica/inmunología , Células B de Memoria/inmunología , Células T de Memoria/inmunología , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Nasofaringe/virología , Nasofaringe/inmunología , Células Plasmáticas/inmunología , Células Plasmáticas/citología , SARS-CoV-2/inmunologíaRESUMEN
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/fisiologíaRESUMEN
INTRODUCTION: The purpose of the study was to analyze the effect of swabs on nasal mucosa. METHODOLOGY: Since May 2020, our department was responsible for screening coronavirus disease 2019 (COVID-19) among the employees of a company that continued its activity during the pandemic. The screening protocol consisted of two swabs per week. The samples were analyzed through objective endoscopic and subjective clinical evaluations with sino-nasal outcome test (SNOT Test) at three time points (T0, T1 - three months, T2 - six months). RESULTS: 23.76% of patients showed an increase in the SNOT score at T1, and the score decreased at T2. This could be due to the phenomenon of "adaptation" of the nasal mucosa. Endoscopic control showed that at T1, secretion, hyperemia, and edema are the most common signs. At T2, however, the crusts accounted for 52.94% of all damage. It is evident that at T1 the endoscopically detected signs of "acute" damage were more represented than at T2, while the signs of "chronic" damage increased as the number of swabs increased. CONCLUSIONS: We demonstrated that mucosal damage and perceived symptoms were absolutely acceptable compared to the diagnostic advantage obtained with serial screening.
Asunto(s)
COVID-19 , Mucosa Nasal , Nasofaringe , SARS-CoV-2 , Humanos , COVID-19/diagnóstico , Mucosa Nasal/virología , SARS-CoV-2/aislamiento & purificación , Masculino , Femenino , Adulto , Persona de Mediana Edad , Nasofaringe/virología , Tamizaje Masivo/métodos , Manejo de Especímenes/métodos , Prueba de COVID-19/métodosRESUMEN
Respiratory syncytial virus is the major cause of acute lower respiratory tract infections in young children, causing extensive mortality and morbidity globally, with limited therapeutic or preventative options. Cathelicidins are innate immune antimicrobial host defence peptides and have antiviral activity against RSV. However, upper respiratory tract cathelicidin expression and the relationship with host and environment factors in early life, are unknown. Infant cohorts were analysed to characterise early life nasal cathelicidin levels, revealing low expression levels in the first week of life, with increased levels at 9 months which are comparable to 2-year-olds and healthy adults. No impact of prematurity on nasal cathelicidin expression was observed, nor were there effects of sex or birth mode, however, nasal cathelicidin expression was lower in the first week-of-life in winter births. Nasal cathelicidin levels were positively associated with specific inflammatory markers and demonstrated to be associated with microbial community composition. Importantly, levels of nasal cathelicidin expression were elevated in infants with mild RSV infection, but, in contrast, were not upregulated in infants hospitalised with severe RSV infection. These data suggest important relationships between nasal cathelicidin, upper airway microbiota, inflammation, and immunity against RSV infection, with interventional potential.
Asunto(s)
Catelicidinas , Infecciones por Virus Sincitial Respiratorio , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Humanos , Femenino , Masculino , Lactante , Recién Nacido , Virus Sincitial Respiratorio Humano/inmunología , Mucosa Nasal/metabolismo , Mucosa Nasal/virología , Mucosa Nasal/inmunologíaRESUMEN
Mucosal immunity may contribute to clearing SARS-CoV-2 infection prior to systemic infection, thereby allowing hosts to remain seronegative. We describe the meaningful detection of SARS-CoV-2-specific nasal mucosal antibodies in a group of exposed-household individuals that evaded systemic infection. Between June 2020 and February 2023, nasopharyngeal swab (NPS) and acute and convalescent blood were collected from individuals exposed to a SARS-CoV-2-confirmed household member. Nasal secretory IgA (SIgA) antibodies targeting the SARS-CoV-2 spike protein were measured using a modified ELISA. Of the 36 exposed individuals without SARS-CoV-2 detected by the RT-PCR of NPS specimens and seronegative for SARS-CoV-2-specific IgG at enrollment and convalescence, 13 (36.1%) had positive SARS-CoV-2-specific SIgA levels detected in the nasal mucosa at enrollment. These individuals had significantly higher nasal SIgA (median 0.52 AU/mL) compared with never-exposed, never-infected controls (0.001 AU/mL) and infected-family participants (0.0002 AU/mL) during the acute visit, respectively (both p < 0.001). The nasal SARS-CoV-2-specific SIgA decreased rapidly over two weeks in the exposed seronegative individuals compared to a rise in SIgA in infected-family members. The nasal SARS-CoV-2-specific SIgA may have a protective role in preventing systemic infection.