Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.790
Filter
1.
Nat Commun ; 15(1): 8355, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333516

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes , COVID-19 , Interferon-gamma , Memory T Cells , Nasal Mucosa , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , Spike Glycoprotein, Coronavirus/immunology , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , SARS-CoV-2/immunology , Nasal Mucosa/immunology , Nasal Mucosa/virology , COVID-19/immunology , COVID-19/virology , Memory T Cells/immunology , Male , Female , Middle Aged , Adult , Integrin alpha1/immunology , Integrin alpha1/metabolism , COVID-19 Vaccines/immunology , Antigens, CD/metabolism , Antigens, CD/immunology , Immunologic Memory/immunology , Integrin alpha Chains
2.
Sci Adv ; 10(39): eado7087, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39331716

ABSTRACT

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.


Subject(s)
Influenza A virus , Interferon Type I , Interleukin-17 , Nasal Mucosa , Orthomyxoviridae Infections , Animals , Female , Pregnancy , Interleukin-17/metabolism , Interleukin-17/immunology , Mice , Interferon Type I/metabolism , Interferon Type I/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Nasal Mucosa/immunology , Nasal Mucosa/virology , Nasal Mucosa/metabolism , Influenza A virus/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Virus Replication , Lung/immunology , Lung/virology
3.
Front Immunol ; 15: 1419527, 2024.
Article in English | MEDLINE | ID: mdl-39286244

ABSTRACT

Mucosal barrier tissues and their mucosal associated lymphoid tissues (MALT) are attractive targets for vaccines and immunotherapies due to their roles in both priming and regulating adaptive immune responses. The upper and lower respiratory mucosae, in particular, possess unique properties: a vast surface area responsible for frontline protection against inhaled pathogens but also simultaneous tight regulation of homeostasis against a continuous backdrop of non-pathogenic antigen exposure. Within the upper and lower respiratory tract, the nasal and bronchial associated lymphoid tissues (NALT and BALT, respectively) are key sites where antigen-specific immune responses are orchestrated against inhaled antigens, serving as critical training grounds for adaptive immunity. Many infectious diseases are transmitted via respiratory mucosal sites, highlighting the need for vaccines that can activate resident frontline immune protection in these tissues to block infection. While traditional parenteral vaccines that are injected tend to elicit weak immunity in mucosal tissues, mucosal vaccines (i.e., that are administered intranasally) are capable of eliciting both systemic and mucosal immunity in tandem by initiating immune responses in the MALT. In contrast, administering antigen to mucosal tissues in the absence of adjuvant or costimulatory signals can instead induce antigen-specific tolerance by exploiting regulatory mechanisms inherent to MALT, holding potential for mucosal immunotherapies to treat autoimmunity. Yet despite being well motivated by mucosal biology, development of both mucosal subunit vaccines and immunotherapies has historically been plagued by poor drug delivery across mucosal barriers, resulting in weak efficacy, short-lived responses, and to-date a lack of clinical translation. Development of engineering strategies that can overcome barriers to mucosal delivery are thus critical for translation of mucosal subunit vaccines and immunotherapies. This review covers engineering strategies to enhance mucosal uptake via active targeting and passive transport mechanisms, with a parallel focus on mechanisms of immune activation and regulation in the respiratory mucosa. By combining engineering strategies for enhanced mucosal delivery with a better understanding of immune mechanisms in the NALT and BALT, we hope to illustrate the potential of these mucosal sites as targets for immunomodulation.


Subject(s)
Immunity, Mucosal , Immunomodulation , Humans , Animals , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Lymphoid Tissue/immunology , Vaccines/immunology , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Administration, Intranasal
4.
Allergol Immunopathol (Madr) ; 52(5): 29-35, 2024.
Article in English | MEDLINE | ID: mdl-39278848

ABSTRACT

Allergic rhinitis (AR) is a chronic, non-infectious inflammatory condition of the nasal mucosa mediated by IgE. There is a need for the development of novel medications to treat this ailment. Isoorientin is a naturally occurring flavonoid that possesses antioxidant, anti--inflammatory, and various other advantageous characteristics. However, its potential effects on AR remain unclear. This study evaluates the therapeutic effects of isoorientin on ovalbumin (OVA)-induced allergic rhinitis (AR) in mice and explores the underlying mechanism. Our study revealed that isoorientin administration effectively decreased the frequency of nose rubbing and sneezing in AR mice. The groups treated with isoorientin showed a significant decrease in serum levels of IgE and histamine, with reductions of 40% and 30%, respectively. Isoorientin ameliorated inflammation of the nasal mucosa and restored the Th1/Th2 balance. In addition, isoorientin inhibited the activation of the NF-κB pathway in nasal tissues. In summary, Isoorientin alleviates OVA-stimulated AR in mice by restoring Th1/Th2 balance and blocking the NF-κB pathway. Thus, isoorientin exhibits promise as a natural therapeutic agent for allergic rhinitis.


Subject(s)
Disease Models, Animal , Immunoglobulin E , Luteolin , Mice, Inbred BALB C , NF-kappa B , Nasal Mucosa , Ovalbumin , Rhinitis, Allergic , Th1-Th2 Balance , Animals , Luteolin/pharmacology , Ovalbumin/immunology , Mice , Rhinitis, Allergic/immunology , Rhinitis, Allergic/drug therapy , Th1-Th2 Balance/drug effects , Nasal Mucosa/immunology , Nasal Mucosa/drug effects , Immunoglobulin E/blood , Immunoglobulin E/immunology , NF-kappa B/metabolism , Th2 Cells/immunology , Female , Humans , Allergens/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Th1 Cells/immunology , Th1 Cells/drug effects , Histamine/metabolism , Histamine/blood
5.
JCI Insight ; 9(18)2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39315545

ABSTRACT

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.


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunity, Mucosal , Immunoglobulin A, Secretory , Adult , Female , Humans , Male , Middle Aged , Adenoviridae , Administration, Intranasal , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Antibodies, Viral/immunology , Antibodies, Viral/blood , COVID-19/prevention & control , COVID-19/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/adverse effects , Genetic Vectors/administration & dosage , Immunoglobulin A, Secretory/immunology , Nasal Mucosa/immunology , Nasal Mucosa/virology , Spike Glycoprotein, Coronavirus/immunology , Vaccination/methods
6.
Inflamm Res ; 73(10): 1819-1832, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39180692

ABSTRACT

PURPOSE: To investigate the immunomodulatory effects and potential mechanisms of human nasal mucosa-derived mesenchymal stem cells(hNMSCs) on mouse allergic rhinitis, and to compare them with human umbilical cord-derived mesenchymal stem cells (hUCMSCs). METHOD: hNMSCs and hUCMSCs were isolated and cultured for identification from human nasal mucosa and umbilical cord tissues. A co-culture system of LPS-stimulated RAW264.7 cells/mouse peritoneal macrophages and MSCs was employed.Changes in inflammatory factors in RAW264.7 cells and the culture medium as well as the expression of NF-κB signaling pathway in RAW264.7 cells were detected. Forty-eight BALB/c mice were randomly divided into control, OVA, hNMSCs, and hUCMSCs groups. An allergic rhinitis (AR) model was established through ovalbumin (OVA) stimulation and treated with hNMSCs and hUCMSCs. Subsequent assessments included related symptoms, biological changes, and the expression of the NF-κB signaling pathway in the nasal mucosa of mice. RESULTS: MSCs can be successfully isolated from human nasal mucosa. Both hNMSCs and hUCMSCs interventions significantly reverseed the inflammation induced by LPS and suppressed the upregulation of the NF-κB signaling pathway in RAW264.7 cells. Treatment with hNMSCs and hUCMSCs alleviated mouse allergic symptoms, reduced levels of total IgE, OVA-specific IgE and IgG1 in mouse serum, TH2-type cytokines and chemokines in mouse nasal mucosa, and TH2-type cytokines in mouse spleen culture medium, while also inhibiting the expression of the NF-κB signaling pathway in the nasal mucosa of mice. moreover, the hNMSCs group showed a more significant reduction in OVA-specific IgG1 in serum and IL-4 expression levels in mouse spleen culture medium compared to the hUCMSCs group. CONCLUSION: Our findings suggest that hNMSCs can ameliorate allergic rhinitis in mice, with a certain advantage in anti-inflammatory effects compared to hUCMSCs. The NF-κB pathway is likely involved in the anti-inflammatory regulation process by hNMSCs.Therefore, hNMSCs might represent a novel therapeutic approach for allergic rhinitis.


Subject(s)
Cytokines , Mesenchymal Stem Cells , Mice, Inbred BALB C , NF-kappa B , Nasal Mucosa , Rhinitis, Allergic , Animals , Nasal Mucosa/immunology , Nasal Mucosa/cytology , Humans , Mesenchymal Stem Cells/immunology , Mice , Rhinitis, Allergic/therapy , Rhinitis, Allergic/immunology , NF-kappa B/metabolism , Cytokines/metabolism , RAW 264.7 Cells , Immunoglobulin E/blood , Female , Lipopolysaccharides/pharmacology , Ovalbumin/immunology , Umbilical Cord/cytology , Coculture Techniques , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Cells, Cultured
7.
Int J Mol Sci ; 25(16)2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39201342

ABSTRACT

Allergic rhinitis (AR) is a series of allergic reactions to allergens in the nasal mucosa and is one of the most common allergic diseases that affect both children and adults. Shi-Bi-Lin (SBL) is the modified formula of Cang Er Zi San (CEZS), a traditional Chinese herbal formula used for treating AR. Our study aims to elucidate the anti-inflammatory effects and mechanisms of SBL in house dust mite-induced AR by regulating gut microflora metabolism. In vivo studies showed that nasal allergies and the infiltration of inflammatory cells in the nasal epithelium were significantly suppressed by SBL. Moreover, SBL restored the impaired nasal epithelial barrier function with an increased tight junction protein expression and reduced the endothelial nitric oxide synthase (eNOS). Interestingly, SBL significantly reconstituted the abundance and composition of gut microbiota in AR mice; it increased the relative abundance of potentially beneficial genera and decreased the relative abundance of harmful genera. SBL also restored immune-related metabolisms, which were significantly increased and correlated with suppressing inflammatory cytokines. Furthermore, a network analysis and molecular docking indicated IL-6 was a possible target drug candidate for the SBL treatment. SBL dramatically reduced the IL-6 level in the nasal lavage fluid (NALF), suppressing the IL-6 downstream Erk1/2 and AKT/PI3K signaling pathways. In conclusion, our study integrates 16S rRNA sequencing, microflora metabolism, and network pharmacology to explain the immune mechanism of SBL in alleviating HDM-induced allergic rhinitis.


Subject(s)
Drugs, Chinese Herbal , Gastrointestinal Microbiome , Network Pharmacology , RNA, Ribosomal, 16S , Rhinitis, Allergic , Rhinitis, Allergic/drug therapy , Rhinitis, Allergic/microbiology , Rhinitis, Allergic/metabolism , Animals , RNA, Ribosomal, 16S/genetics , Mice , Drugs, Chinese Herbal/pharmacology , Gastrointestinal Microbiome/drug effects , Nasal Mucosa/metabolism , Nasal Mucosa/microbiology , Nasal Mucosa/drug effects , Nasal Mucosa/immunology , Pyroglyphidae/immunology , Molecular Docking Simulation , Disease Models, Animal , Mice, Inbred BALB C , Female
8.
Nano Lett ; 24(33): 10380-10387, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39120059

ABSTRACT

The advancement of effective nasal mucoadhesive delivery faces challenges due to rapid mucociliary clearance (MCC). Conventional studies have employed mucoadhesive materials, mainly forming spherical nanoparticles, but these offer limited adhesion to the nasal mucosa. This study hypothesizes that a 2D nanoscale structure utilizing adhesive polyphenols can provide a superior strategy for countering MCC, aligning with the planar mucosal layers. We explore the use of tannic acid (TA), a polyphenolic molecule known for its adhesive properties and ability to form complexes with biomolecules. Our study introduces an unprecedented 2D nanopatch, assembled through the interaction of TA with green fluorescent protein (GFP), and cell-penetrating peptide (CPP). This 2D nanopatch demonstrates robust adhesion to nasal mucosa and significantly enhances immunoglobulin A secretions, suggesting its potential for enhancing nasal vaccine delivery. The promise of a polyphenol-enabled adhesive 2D nanopatch signifies a pivotal shift from conventional spherical nanoparticles, opening new pathways for delivery strategies through respiratory mucoadhesion.


Subject(s)
Nasal Mucosa , Polyphenols , Tannins , Tannins/chemistry , Polyphenols/chemistry , Polyphenols/administration & dosage , Nasal Mucosa/metabolism , Nasal Mucosa/immunology , Animals , Nanoparticles/chemistry , Humans , Cell-Penetrating Peptides/chemistry , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/chemistry , Adhesives/chemistry , Mucociliary Clearance/drug effects , Immunoglobulin A , Mice
9.
Front Immunol ; 15: 1394539, 2024.
Article in English | MEDLINE | ID: mdl-39176088

ABSTRACT

There is growing evidence that neurogenic inflammation contributes to the pathophysiology of upper airway diseases, with nasal hyperreactivity (NHR) being a key symptom. The rare neuroendocrine cells (NECs) in the epithelium have been linked to the pathophysiology of bronchial and intestinal hyperreactivity, however their presence in the nasal mucosa and their potential role in NHR remains unclear. Therefore, we studied the presence of NECs in the nasal epithelium of controls, allergic rhinitis patients and chronic rhinosinusitis with nasal polyps patients, and their link to NHR. The expression of typical NECs markers, CHGA, ASCL1 and CGRP, were evaluated on gene and protein level in human samples using real-time quantitative PCR (RT-qPCR), western blot, immunohistochemistry fluorescence staining, RNA scope assay, flow cytometry and single cell RNA-sequencing. Furthermore, the change in peak nasal inspiratory flow after cold dry air provocation and visual analogue scale scores were used to evaluate NHR or disease severity, respectively. Limited gene expression of the NECs markers CHGA and ASCL1 was measured in patients with upper airway diseases and controls. Gene expression of these markers did not correlate with NHR severity nor disease severity. In vitro, CHGA and ASCL1 expression was also evaluated in primary nasal epithelial cell cultures from patients with upper airway disease and controls using RT-qPCR and western blot. Both on gene and protein level only limited CHGA and ASCL1 expression was found. Additionally, NECs were studied in nasal biopsies of patients with upper airway diseases and controls using immunohistochemistry fluorescence staining, RNA scope and flow cytometry. Unlike in ileum samples, CHGA could not be detected in nasal biopsies of patients with upper airway diseases and control subjects. Lastly, single cell RNA-sequencing of upper airway tissue could not identify a NEC cluster. In summary, in contrast to the bronchi and gut, there is only limited evidence for the presence of NECs in the nasal mucosa, and without correlation with NHR, thereby questioning the relevance of NECs in upper airway pathology.


Subject(s)
Nasal Mucosa , Nasal Polyps , Neuroendocrine Cells , Humans , Nasal Mucosa/metabolism , Nasal Mucosa/pathology , Nasal Mucosa/immunology , Female , Adult , Male , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Middle Aged , Nasal Polyps/immunology , Nasal Polyps/pathology , Nasal Polyps/metabolism , Sinusitis/metabolism , Sinusitis/pathology , Sinusitis/immunology , Rhinitis, Allergic/metabolism , Rhinitis, Allergic/immunology , Rhinitis, Allergic/pathology , Biomarkers , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured
10.
Front Immunol ; 15: 1430760, 2024.
Article in English | MEDLINE | ID: mdl-39185421

ABSTRACT

Allergic rhinitis (AR) is a chronic, non-infectious condition affecting the nasal mucosa, primarily mediated mainly by IgE. Recent studies reveal that AR is intricately associated not only with type 2 immunity but also with neuroimmunity. Nociceptive neurons, a subset of primary sensory neurons, are pivotal in detecting external nociceptive stimuli and modulating immune responses. This review examines nociceptive neuron receptors and elucidates how neuropeptides released by these neurons impact the immune system. Additionally, we summarize the role of immune cells and inflammatory mediators on nociceptive neurons. A comprehensive understanding of the dynamic interplay between nociceptive neurons and the immune system augments our understanding of the neuroimmune mechanisms underlying AR, thereby opening novel avenues for AR treatment modalities.


Subject(s)
Nociceptors , Rhinitis, Allergic , Humans , Nociceptors/metabolism , Nociceptors/immunology , Rhinitis, Allergic/immunology , Rhinitis, Allergic/metabolism , Animals , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Nasal Mucosa/innervation , Neuroimmunomodulation , Neuropeptides/metabolism , Neuropeptides/immunology
11.
Sci Adv ; 10(34): eadp2636, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39178263

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , COVID-19 , Mice, Inbred C57BL , SARS-CoV-2 , Virus Replication , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , SARS-CoV-2/physiology , SARS-CoV-2/immunology , COVID-19/virology , COVID-19/immunology , COVID-19/prevention & control , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/metabolism , Mice , Lung/virology , Lung/immunology , Humans , Female , Nasal Mucosa/virology , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Granzymes/metabolism
12.
Signal Transduct Target Ther ; 9(1): 190, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39039046

ABSTRACT

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.


Subject(s)
Administration, Intranasal , Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , SARS-CoV-2/immunology , SARS-CoV-2/genetics , COVID-19/prevention & control , COVID-19/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Animals , Mice , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Viral/immunology , Antibodies, Viral/blood , COVID-19 Vaccines/immunology , COVID-19 Vaccines/genetics , COVID-19 Vaccines/administration & dosage , Immunoglobulin A/immunology , Immunoglobulin A/blood , Immunoglobulin A/genetics , Nasal Mucosa/immunology , Nasal Mucosa/virology , Female , Genetic Vectors/immunology , Genetic Vectors/genetics , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/immunology , Immunoglobulin A, Secretory/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Immunoglobulin G/immunology , Immunoglobulin G/blood , Male
13.
Front Immunol ; 15: 1429442, 2024.
Article in English | MEDLINE | ID: mdl-39040099

ABSTRACT

Introduction: Allergic rhinitis (AR) is an upper airway inflammatory disease of the nasal mucosa. Conventional treatments such as symptomatic pharmacotherapy and allergen-specific immunotherapy have considerable limitations and drawbacks. As an emerging therapy with regenerative potential and immunomodulatory effect, mesenchymal stem cell-derived exosomes (MSC-Exos) have recently been trialed for the treatment of various inflammatory and autoimmune diseases. Methods: In order to achieve sustained and protected release of MSC-Exos for intranasal administration, we fabricated Poly(lactic-co-glycolic acid) (PLGA) micro and nanoparticles-encapsulated MSC-Exos (PLGA-Exos) using mechanical double emulsion for local treatment of AR. Preclinical in vivo imaging, ELISA, qPCR, flow cytometry, immunohistochemical staining, and multiomics sequencing were used for phenotypic and mechanistic evaluation of the therapeutic effect of PLGA-Exos in vitro and in vivo. Results: The results showed that our PLGA platform could efficiently encapsulate and release the exosomes in a sustained manner. At protein level, PLGA-Exos treatment upregulated IL-2, IL-10 and IFN-γ, and downregulated IL-4, IL-17 and antigen-specific IgE in ovalbumin (OVA)-induced AR mice. At cellular level, exosomes treatment reduced Th2 cells, increased Tregs, and reestablished Th1/Th2 balance. At tissue level, PLGA-Exos significantly attenuated the infiltration of immune cells (e.g., eosinophils and goblet cells) in nasal mucosa. Finally, multiomics analysis discovered several signaling cascades, e.g., peroxisome proliferator-activated receptor (PPAR) pathway and glycolysis pathway, that might mechanistically support the immunomodulatory effect of PLGA-Exos. Discussion: For the first time, we present a biomaterial-facilitated local delivery system for stem cell-derived exosomes as a novel and promising strategy for AR treatment.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Polylactic Acid-Polyglycolic Acid Copolymer , Rhinitis, Allergic , Exosomes/immunology , Exosomes/metabolism , Animals , Rhinitis, Allergic/therapy , Rhinitis, Allergic/immunology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Immunomodulation , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Administration, Intranasal
14.
J Ethnopharmacol ; 335: 118611, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39053712

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Allergic rhinitis (AR) stands as a non-infectious inflammatory condition affecting the nasal mucosa, marked by bouts of sneezing, nasal itching, and congestion. This ailment afflicts individuals across all age groups and poses challenges for effective treatment due to its chronic nature. Cangerzisan (CEZS), documented in the Jishengfang compendium, represents a traditional Chinese medicinal formula long utilized for AR management. AIM OF THE STUDY: Investigating mechanism beneath therapeutic effect of CEZS in alleviating AR. MATERIALS AND METHODS: The main active components in CEZS were determined by High Performance Liquid Chromatography (HPLC).The active constituents of CEZS and their corresponding targets were identified through an exhaustive screening process employing TCMSP database. To identify targets relevant to AR, GeneCards, OMIM, and DisGeNET databases were thoroughly applied. Protein-protein interaction (PPI) network was assembled utilizing STRING platform. Potential signaling pathways influenced by CEZS were delineated through GO and KEGG enrichment analyses. Subsequently, an AR model was induced by administering aluminum hydroxide (Al(OH)3) and ovalbumin (OVA) for affecting basal and local sensitization, respectively, facilitating experimental validation of the principal signaling pathways. RESULTS: There were 61 active constituents identified within CEZS, targeting a pool of 129 entities associated with AR treatment. Pathways analysis of KEGG revealed that CEZS potentially inhibits AR advancement via modulating TLR4 signaling pathway. Animal experiments demonstrated that CEZS effectively alleviated symptom scores in guinea pigs with AR. Moreover, it exhibited notable improvements in serum immune and inflammatory factors levels, as well as reduced inflammatory infiltration within nasal mucosa, including goblet and mast cells. CEZS was found to enhance GATA-3 expression while reducing T-bet expression, thereby modulating the TH1/TH2 immune balance. Additionally, CEZS downregulated HMGB1, TLR4, and p-NF-κB/NF-κB protein expressions within nasal mucosa of guinea pigs. CONCLUSIONS: The therapeutic mechanism of CEZS against AR involves rectifying TH1/TH2 immune imbalance and upregulating inflammatory and immune factors through modulating key proteins expression within TLR4 pathway. This targeted regulation effectively impedes AR progression.


Subject(s)
Network Pharmacology , Rhinitis, Allergic , Animals , Rhinitis, Allergic/drug therapy , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/chemistry , Protein Interaction Maps , Signal Transduction/drug effects , Ovalbumin , Male , Female , Toll-Like Receptor 4/metabolism , Disease Models, Animal , Nasal Mucosa/drug effects , Nasal Mucosa/metabolism , Nasal Mucosa/immunology , Guinea Pigs , Anti-Allergic Agents/pharmacology , Cytokines/metabolism
15.
HNO ; 72(9): 633-638, 2024 Sep.
Article in German | MEDLINE | ID: mdl-39031180

ABSTRACT

Diagnosis of allergic disease is primarily verified by IgE blood serum analysis. Determination in nasal secretions is technically more difficult, particularly due to a low specimen volume and the method of sample collection. Nasal secretions are frequently collected by lavage, which allows qualitative diagnostics, whereas swabs with defined amounts of mucus enable quantitative analyses. In the case of negative skin and serum tests, detection of IgE in nasal mucus combined with nasal provocation testing aids differentiation between local allergic and nonallergic rhinitis.


Subject(s)
Immunoglobulin E , Nasal Mucosa , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Nasal Mucosa/immunology , Nasal Mucosa/metabolism
16.
Nature ; 632(8025): 637-646, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39085603

ABSTRACT

Nasal vaccination elicits a humoral immune response that provides protection from airborne pathogens1, yet the origins and specific immune niches of antigen-specific IgA-secreting cells in the upper airways are unclear2. Here we define nasal glandular acinar structures and the turbinates as immunological niches that recruit IgA-secreting plasma cells from the nasal-associated lymphoid tissues (NALTs)3. Using intact organ imaging, we demonstrate that nasal vaccination induces B cell expansion in the subepithelial dome of the NALT, followed by invasion into commensal-bacteria-driven chronic germinal centres in a T cell-dependent manner. Initiation of the germinal centre response in the NALT requires pre-expansion of antigen-specific T cells, which interact with cognate B cells in interfollicular regions. NALT ablation and blockade of PSGL-1, which mediates interactions with endothelial cell selectins, demonstrated that NALT-derived IgA-expressing B cells home to the turbinate region through the circulation, where they are positioned primarily around glandular acinar structures. CCL28 expression was increased in the turbinates in response to vaccination and promoted homing of IgA+ B cells to this site. Thus, in response to nasal vaccination, the glandular acini and turbinates provide immunological niches that host NALT-derived IgA-secreting cells. These cellular events could be manipulated in vaccine design or in the treatment of upper airway allergic responses.


Subject(s)
Immunoglobulin A , Lymphoid Tissue , Nasal Mucosa , Plasma Cells , T-Lymphocytes , Turbinates , Animals , Female , Male , Mice , Bacteria/immunology , Cell Movement , Chemokines, CC/immunology , Chemokines, CC/metabolism , Germinal Center/immunology , Germinal Center/cytology , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Lymphoid Tissue/immunology , Lymphoid Tissue/cytology , Mice, Inbred C57BL , Nasal Mucosa/cytology , Nasal Mucosa/immunology , Plasma Cells/immunology , Plasma Cells/cytology , Plasma Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Turbinates/cytology , Turbinates/immunology , Vaccination , Administration, Intranasal , Vaccines/immunology , Symbiosis
17.
Nature ; 632(8025): 630-636, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39085605

ABSTRACT

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.


Subject(s)
Immunologic Memory , Memory B Cells , Memory T Cells , Nasal Mucosa , Nasopharynx , SARS-CoV-2 , Adult , Humans , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , COVID-19/immunology , COVID-19/virology , Germinal Center/immunology , Germinal Center/cytology , Immunoglobulin A/immunology , Immunologic Memory/immunology , Memory B Cells/immunology , Memory T Cells/immunology , Nasal Mucosa/immunology , Nasal Mucosa/virology , Nasopharynx/virology , Nasopharynx/immunology , Plasma Cells/immunology , Plasma Cells/cytology , SARS-CoV-2/immunology
18.
J Exp Med ; 221(9)2024 09 02.
Article in English | MEDLINE | ID: mdl-38949639

ABSTRACT

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.


Subject(s)
COVID-19 , Immunity, Innate , Nasal Mucosa , SARS-CoV-2 , Humans , COVID-19/immunology , Child , Immunity, Innate/immunology , SARS-CoV-2/immunology , Nasal Mucosa/virology , Nasal Mucosa/immunology
19.
J Exp Med ; 221(9)2024 09 02.
Article in English | MEDLINE | ID: mdl-38949638

ABSTRACT

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.


Subject(s)
COVID-19 , Immunity, Innate , SARS-CoV-2 , Humans , Immunity, Innate/immunology , Child, Preschool , Infant , COVID-19/immunology , COVID-19/virology , Child , SARS-CoV-2/immunology , Female , Male , Nasopharynx/immunology , Nasopharynx/virology , Nasopharynx/microbiology , Viral Load , Nasal Mucosa/immunology , Nasal Mucosa/virology , Nasal Mucosa/microbiology , Cytokines/metabolism , Cytokines/immunology , Host-Pathogen Interactions/immunology , Adolescent , Nose/immunology , Nose/virology , Nose/microbiology , Coinfection/immunology , Coinfection/virology
20.
Immunity ; 57(8): 1955-1974.e8, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38964332

ABSTRACT

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.


Subject(s)
Immunologic Memory , Memory T Cells , Nasal Mucosa , Orthomyxoviridae Infections , Animals , Immunologic Memory/immunology , Mice , Nasal Mucosa/virology , Nasal Mucosa/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Memory T Cells/immunology , Epithelial Cells/immunology , Epithelial Cells/virology , Mice, Inbred C57BL , Humans , Single-Cell Analysis , Influenza, Human/immunology , Influenza, Human/virology , Female , Receptors, CXCR6/metabolism , Receptors, CXCR6/immunology , Influenza A virus/immunology , Influenza A virus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL