Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.556
Filter
1.
Acta Med Okayama ; 78(2): 95-106, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38688827

ABSTRACT

The lungs are very complex organs, and the respiratory system performs the dual roles of repairing tissue while protecting against infection from various environmental stimuli. Persistent external irritation disrupts the immune responses of tissues and cells in the respiratory system, ultimately leading to respiratory disease. Neuropeptide Y (NPY) is a 36-amino-acid polypeptide and a neurotransmitter that regulates homeostasis. The NPY receptor is a seven-transmembrane-domain G-protein-coupled receptor with six subtypes (Y1, Y2, Y3, Y4, Y5, and Y6). Of these receptors, Y1, Y2, Y4, and Y5 are functional in humans, and Y1 plays important roles in the immune responses of many organs, including the respiratory system. NPY and the Y1 receptor have critical roles in the pathogenesis of asthma, chronic obstructive pulmonary disease, and idiopathic pulmonary fibrosis. The effects of NPY on the airway immune response and pathogenesis differ among respiratory diseases. This review focuses on the involvement of NPY in the airway immune response and pathogenesis of various respiratory diseases.


Subject(s)
Neuropeptide Y , Receptors, Neuropeptide Y , Humans , Neuropeptide Y/physiology , Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/physiology , Animals , Respiratory Tract Diseases/immunology , Asthma/immunology , Respiratory System/immunology , Pulmonary Disease, Chronic Obstructive/immunology
2.
Nature ; 626(7998): 392-400, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38086420

ABSTRACT

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


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Memory T Cells , Paramyxoviridae Infections , Respiratory System , Animals , Mice , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/metabolism , Immunity, Herd/immunology , Immunologic Memory/immunology , Interferon-gamma/immunology , Memory T Cells/immunology , Paramyxoviridae/immunology , Paramyxoviridae/physiology , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/prevention & control , Paramyxoviridae Infections/transmission , Paramyxoviridae Infections/virology , Respiratory System/cytology , Respiratory System/immunology , Respiratory System/virology , Transcription, Genetic , Humans
4.
Sci Rep ; 13(1): 7162, 2023 05 03.
Article in English | MEDLINE | ID: mdl-37138005

ABSTRACT

Coronavirus disease (COVID-19) has generated interest in the assessment of systemic immune status, but existing knowledge about mucosal immunity is clearly insufficient to understand the full pathogenetic mechanisms of the disease. The aim of this study was to evaluate the long-term effects of novel coronavirus infection on mucosal immunity in the postinfection period among health care workers (HCWs). A total of 180 health care workers with and without a history of COVID-19 who ranged in age from 18 to 65 years were enrolled in this one-stage, cross-sectional study. The study subjects completed the 36-Item Short Form (36) Health Survey (SF-36) and the Fatigue Assessment Scale. Secretory immunoglobulin A (sIgA) and total immunoglobulin G (IgG) levels were quantified in saliva samples, induced sputum samples, and nasopharyngeal and oropharyngeal scrapings by an enzyme-linked immunosorbent assay. Specific anti-SARS-CoV-2 IgG antibodies were quantified in serum samples by chemiluminescence immunoassay. Analysis of the questionnaire data showed that all HCWs with a history of COVID-19 reported health problems that limited their daily activities and negative changes in their emotional health three months after the disease, regardless of its severity. The following shifts were detected in the adaptive arm of the immune response in different mucosal compartments. Among subjects who had severe or moderate-to-severe COVID-19, salivary sIgA levels were significantly higher than those in the control group (p < 0.05 and p < 0.005, respectively). Compared to the subjects in the control group, all subjects with prior COVID-19 had significantly higher levels of total IgG in induced sputum. In the group of patients who had had severe infection, total IgG in saliva was also higher (p < 0.05). A direct statistically significant correlation was also detected between the levels of total IgG in all studied samples and the levels of specific IgG antibodies against SARS-CoV-2 in the serum. A significant correlation was observed between total IgG levels and the parameters of physical and social activities, mental health, and fatigue levels. Our study demonstrated long-term changes in the humoral mucosal immune response, which were most pronounced in health care workers with a history of severe or moderate-to-severe COVID-19, and an association of these changes with certain clinical signs of post-COVID-19 syndrome.


Subject(s)
COVID-19 , Health Personnel , Immunity, Mucosal , Russia , COVID-19/immunology , COVID-19/pathology , COVID-19/physiopathology , Humans , Young Adult , Adult , Middle Aged , Immunoglobulin A/analysis , Respiratory System/immunology , Antibodies, Viral/analysis , Severity of Illness Index , Immunoglobulin G/analysis , SARS-CoV-2/physiology
5.
Viruses ; 15(2)2023 01 31.
Article in English | MEDLINE | ID: mdl-36851616

ABSTRACT

Type 2 immune responses are characterized by elevated type 2 cytokines and blood eosinophilia. Emerging evidence suggests that people with chronic type 2 inflammatory lung diseases are not particularly susceptible to SARS-CoV-2 infection. Intriguingly, recent in vitro, ex vivo research demonstrates type 2 cytokines, particularly IL-13, reduce the risk of SARS-CoV-2 infection in the airway epithelium. IL-13 treatment in airway epithelial cells followed by SARS-CoV-2 diminished viral entry, replication, spread, and cell death. IL-13 reduces the expression of the angiotensin-converting enzyme 2 (ACE2) receptor in the airway epithelium and transmembrane serine protease 2 (TMPRSS2), particularly in ciliated cells. It also alters the cellular composition toward a secretory-cell-rich phenotype reducing total ciliated cells and, thus, reducing viral tropism. IL-13 enhances Muc5ac mucin and glycocalyx secretion in the periciliary layer, which acts as a physical barrier to restrict virus attachment. Moreover, type 2 airway immune cells, such as M2 alveolar macrophages, CD4+ tissue-resident memory T cells, and innate lymphoid 2 cells, may also rescue type 2 airways from SARS-CoV-2-induced adverse effects. In this review, we discuss recent findings that demonstrate how type 2 immunity alters immune responses against SARS-CoV-2 and its consequences on COVID-19 pathogenesis.


Subject(s)
COVID-19 , Humans , Cytokines , Immunity, Innate , Interleukin-13 , Lymphocytes , SARS-CoV-2 , Respiratory System/immunology
6.
Clin Rev Allergy Immunol ; 64(2): 161-178, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35275333

ABSTRACT

The respiratory tract is home to a diverse microbial community whose influence on local and systemic immune responses is only beginning to be appreciated. Increasing reports have linked changes in this microbiome to a range of pulmonary and extrapulmonary disorders, including asthma, chronic obstructive pulmonary disease and rheumatoid arthritis. Central to many of these findings is the role of IL-17-type immunity as an important driver of inflammation. Despite the crucial role played by IL-17-mediated immune responses in protection against infection, overt Th17 cell responses have been implicated in the pathogenesis of several chronic inflammatory diseases. However, our knowledge of the influence of bacteria that commonly colonise the respiratory tract on IL-17-driven inflammatory responses remains sparse. In this article, we review the current knowledge on the role of specific members of the airway microbiota in the modulation of IL-17-type immunity and discuss how this line of research may support the testing of susceptible individuals and targeting of inflammation at its earliest stages in the hope of preventing the development of chronic disease.


Subject(s)
Inflammation , Interleukin-17 , Lung , Microbiota , Humans , Chronic Disease , Inflammation/immunology , Interleukin-17/immunology , Lung/immunology , Lung/microbiology , Microbiota/immunology , Respiratory System/immunology , Respiratory System/microbiology
7.
Neumol. pediátr. (En línea) ; 18(1): 6-8, 2023. tab, ilus
Article in Spanish | LILACS | ID: biblio-1442597

ABSTRACT

El sistema respiratorio cumple múltiples funciones no respiratorias, entre ellas es fundamental su rol en la defensa del organismo frente a una gran variedad de agentes externos potencialmente nocivos. Para ello, cuenta con mecanismos de protección de la vía aérea, dado por sus estructuras anatómicas, reflejos y el transporte mucociliar, además de un complejo y amplio desarrollo del sistema inmune pulmonar. El objetivo de este articulo es revisar los conceptos y componentes más importantes de función defensiva del sistema respiratorio.


The respiratory system fulfills multiple non-respiratory functions, including its role in defending the body against a wide variety of potentially harmful external agents. For this, it has airway protection mechanisms, given by its anatomical structures, reflexes and mucociliary transport, as well as a complex and extensive development of the pulmonary immune system. The objective of this article is to review the most important concepts and components of the defensive function of the respiratory system.


Subject(s)
Humans , Respiratory System/immunology
8.
Proc Natl Acad Sci U S A ; 119(24): e2202069119, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35679343

ABSTRACT

Current vaccines have greatly diminished the severity of the COVID-19 pandemic, even though they do not entirely prevent infection and transmission, likely due to insufficient immunity in the upper respiratory tract. Here, we compare intramuscular and intranasal administration of a live, replication-deficient modified vaccinia virus Ankara (MVA)-based Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) spike (S) vaccine to raise protective immune responses in the K18-hACE2 mouse model. Using a recombinant MVA expressing firefly luciferase for tracking, live imaging revealed luminescence of the respiratory tract of mice within 6 h and persisting for 3 d following intranasal inoculation, whereas luminescence remained at the site of intramuscular vaccination. Intramuscular vaccination induced S-binding-Immunoglobulin G (IgG) and neutralizing antibodies in the lungs, whereas intranasal vaccination also induced Immunoglobulin A (IgA) and higher levels of antigen-specific CD3+CD8+IFN-γ+ T cells. Similarly, IgG and neutralizing antibodies were present in the blood of mice immunized intranasally and intramuscularly, but IgA was detected only after intranasal inoculation. Intranasal boosting increased IgA after intranasal or intramuscular priming. While intramuscular vaccination prevented morbidity and cleared SARS-CoV-2 from the respiratory tract within several days after challenge, intranasal vaccination was more effective as neither infectious virus nor viral messenger (m)RNAs were detected in the nasal turbinates or lungs as early as 2 d after challenge, indicating prevention or rapid elimination of SARS-CoV-2 infection. Additionally, we determined that neutralizing antibody persisted for more than 6 mo and that serum induced to the Wuhan S protein neutralized pseudoviruses expressing the S proteins of variants, although with less potency, particularly for Beta and Omicron.


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunoglobulin A , Respiratory System , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Vaccinia virus , Administration, Intranasal , Angiotensin-Converting Enzyme 2/genetics , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19/prevention & control , COVID-19/transmission , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Mice , Mice, Transgenic , Respiratory System/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccination/methods , Vaccinia virus/genetics , Vaccinia virus/immunology
9.
J Virol ; 96(14): e0073822, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35762755

ABSTRACT

Respiratory coronaviruses cause serious health threats to humans and animals. Porcine respiratory coronavirus (PRCoV), a natural transmissible gastroenteritis virus (TGEV) mutant with partial spike deletion, causes mild respiratory disease and is an interesting animal respiratory coronavirus model for human respiratory coronaviruses. However, the absence of robust ex vivo models of porcine airway epithelium hinders an understanding of the pathogenesis of PRCoV infection. Here, we generated long-term porcine airway organoids (AOs) derived from basal epithelial cells, which recapitulate the in vivo airway complicated epithelial cellularity. Both 3D and 2D AOs are permissive for PRCoV infection. Unlike TGEV, which established successful infection in both AOs and intestinal organoids, PRCoV was strongly amplified only in AOs, not intestinal organoids. Furthermore, PRCoV infection in AOs mounted vigorous early type I and III interferon (IFN) responses and upregulated the expression of overzealous inflammatory genes, including pattern recognition receptors (PRRs) and proinflammatory cytokines. Collectively, these data demonstrate that stem-derived porcine AOs can serve as a promising disease model for PRCoV infection and provide a valuable tool to study porcine respiratory infection. IMPORTANCE Porcine respiratory CoV (PRCoV), a natural mutant of TGEV, shows striking pathogenetic similarities to human respiratory CoV infection and provides an interesting animal model for human respiratory CoVs, including SARS-CoV-2. The lack of an in vitro model recapitulating the complicated cellularity and structure of the porcine respiratory tract is a major roadblock for the study of PRCoV infection. Here, we developed long-term 3D airway organoids (AOs) and further established 2D AO monolayer cultures. The resultant 3D and 2D AOs are permissive for PRCoV infection. Notably, PRCoV mediated pronounced IFN and inflammatory responses in AOs, which recapitulated the inflammatory responses associated with PRCoV in vivo infection. Therefore, porcine AOs can be utilized to characterize the pathogenesis of PRCoV and, more broadly, can serve as a universal platform for porcine respiratory infection.


Subject(s)
Immunity, Innate , Organoids , Porcine Respiratory Coronavirus , Respiratory System , Animals , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/physiopathology , Disease Models, Animal , Humans , Organoids/immunology , Organoids/virology , Respiratory System/immunology , Respiratory System/virology , SARS-CoV-2 , Swine
10.
Bol. latinoam. Caribe plantas med. aromát ; 21(2): 176-206, mar. 2022. ilus, tab
Article in English | LILACS | ID: biblio-1393396

ABSTRACT

Currently, the whole world is facing a life-threatening novel coronavirus 2019 (COVID-19) pandemic. Natural products are well-known for their potential role against viral disease, and some anti-viral agents have been developed to combat these diseases. Herein, the authors investigated the possible effects of this Holy plant Nigella sativa L. (NS), against coronavirus, using evidence-based and mechanistic approaches to conclude the immune-boosting and alleviation of respiratory systemeffects of NS. The pharmacological studies established a prominent role in treating various respiratory, immune systems, cardiovascular, skin, and gastrointestinal disorders. Literature supported the significant anti-viral role and showed an inhibitory role for NS against MHV-A59 CoV (mouse-hepatitis virus­A59) infected Hela, i.e., HeLaCEACAM1a (HeLa-epithelial carcinoembryonic antigen-related cell adhesion molecule 1a) cell. NS is a safe herbal product or dietary supplement and could be an effective and affordable community adjuvant treatment for coronavirus in the current scenario.


Actualmente, el mundo entero se enfrenta a una pandemia del nuevo coronavirus 2019 (COVID-19) que amenaza la vida. Los productos naturales son bien conocidos por su papel potencial contra las enfermedades virales, y se han desarrollado algunos agentes antivirales para combatir estas enfermedades. En este documento, los autores investigaron los posibles efectos de esta planta sagrada Nigella sativa L. (NS), contra el coronavirus, utilizando enfoques mecanicistas y basados en la evidencia para concluir el refuerzo inmunológico y el alivio de los efectos del SN en el sistema respiratorio. Los estudios farmacológicos establecieron un papel destacado en el tratamiento de diversos trastornos respiratorios, del sistema inmunológico, cardiovasculares, cutáneos y gastrointestinales. La literatura apoyó el importante papel antivírico y mostró un papel inhibidor de NS contra células Hela infectadas con MHV-A59 CoV (virus de la hepatitis de ratón-A59), es decir, HeLaCEACAM1a (molécula de adhesión celular 1a relacionada con el antígeno carcinoembrionario epitelial de HeLa). NS es un producto a base de hierbas o un suplemento dietético seguro y podría ser un tratamiento adyuvante comunitario eficaz y asequible para el coronavirus en el escenario actual.


Subject(s)
Humans , Antiviral Agents/pharmacology , Plant Extracts/pharmacology , Nigella sativa/chemistry , COVID-19/drug therapy , Antiviral Agents/immunology , Respiratory System/drug effects , Respiratory System/immunology , Plant Extracts/immunology , Anti-Asthmatic Agents , COVID-19/immunology , Immune System/drug effects
11.
Microbiome ; 10(1): 34, 2022 02 21.
Article in English | MEDLINE | ID: mdl-35189979

ABSTRACT

BACKGROUND: There is increasing evidence that the airway microbiome plays a key role in the establishment of respiratory health by interacting with the developing immune system early in life. While it has become clear that bacteria are involved in this process, there is a knowledge gap concerning the role of fungi. Moreover, the inter-kingdom interactions that influence immune development remain unknown. In this prospective exploratory human study, we aimed to determine early post-natal microbial and immunological features of the upper airways in 121 healthy newborns. RESULTS: We found that the oropharynx and nasal cavity represent distinct ecological niches for bacteria and fungi. Breastfeeding correlated with changes in microbiota composition of oropharyngeal samples with the greatest impact upon the relative abundance of Streptococcus species and Candida. Host transcriptome profiling revealed that genes with the highest expression variation were immunological in nature. Multi-omics factor analysis of host and microbial data revealed unique co-variation patterns. CONCLUSION: These data provide evidence of a diverse multi-kingdom microbiota linked with local immunological characteristics in the first week of life that could represent distinct trajectories for future respiratory health. TRIAL REGISTRATION: NHS Health Research Authority, IRAS ID 199053. Registered 5 Oct 2016. https://www.hra.nhs.uk/planning-and-improving-research/application-summaries/research-summaries/breathing-together/ Video abstract.


Subject(s)
Microbiota , Bacteria , Humans , Infant, Newborn , Microbiota/genetics , Microbiota/immunology , Oropharynx/immunology , Oropharynx/microbiology , Prospective Studies , Respiratory System/immunology , Respiratory System/microbiology
12.
Immunity ; 55(3): 542-556.e5, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35151371

ABSTRACT

Some patients hospitalized with acute COVID-19 suffer respiratory symptoms that persist for many months. We delineated the immune-proteomic landscape in the airways and peripheral blood of healthy controls and post-COVID-19 patients 3 to 6 months after hospital discharge. Post-COVID-19 patients showed abnormal airway (but not plasma) proteomes, with an elevated concentration of proteins associated with apoptosis, tissue repair, and epithelial injury versus healthy individuals. Increased numbers of cytotoxic lymphocytes were observed in individuals with greater airway dysfunction, while increased B cell numbers and altered monocyte subsets were associated with more widespread lung abnormalities. A one-year follow-up of some post-COVID-19 patients indicated that these abnormalities resolved over time. In summary, COVID-19 causes a prolonged change to the airway immune landscape in those with persistent lung disease, with evidence of cell death and tissue repair linked to the ongoing activation of cytotoxic T cells.


Subject(s)
B-Lymphocytes/immunology , COVID-19/immunology , Monocytes/immunology , Respiration Disorders/immunology , Respiratory System/immunology , SARS-CoV-2/physiology , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , COVID-19/complications , Female , Follow-Up Studies , Humans , Immunity, Cellular , Immunoproteins , Male , Middle Aged , Proteome , Respiration Disorders/etiology , Respiratory System/pathology
13.
mSphere ; 7(1): e0098421, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35044807

ABSTRACT

Streptococcus pneumoniae (the pneumococcus) is a leading cause of pneumonia in children under 5 years of age. Coinfection by pneumococci and respiratory viruses enhances disease severity. Little is known about pneumococcal coinfections with respiratory syncytial virus (RSV). Here, we developed a novel infant mouse model of coinfection using pneumonia virus of mice (PVM), a murine analogue of RSV, to examine the dynamics of coinfection in the upper respiratory tract, an anatomical niche that is essential for host-to-host transmission and progression to disease. Coinfection increased damage to the nasal tissue and increased production of the chemokine CCL3. Nasopharyngeal pneumococcal density and shedding in nasal secretions were increased by coinfection. In contrast, coinfection reduced PVM loads in the nasopharynx, an effect that was independent of pneumococcal strain and the order of infection. We showed that this "antagonistic" effect was absent using either ethanol-killed pneumococci or a pneumococcal mutant deficient in capsule production and incapable of nasopharyngeal carriage. Colonization with a pneumococcal strain naturally unable to produce capsule also reduced viral loads. The pneumococcus-mediated reduction in PVM loads was caused by accelerated viral clearance from the nasopharynx. Although these synergistic and antagonistic effects occurred with both wild-type pneumococcal strains used in this study, the magnitude of the effects was strain dependent. Lastly, we showed that pneumococci can also antagonize influenza virus. Taken together, our study has uncovered multiple novel facets of bacterial-viral coinfection. Our findings have important public health implications, including for bacterial and viral vaccination strategies in young children. IMPORTANCE Respiratory bacterial-viral coinfections (such as pneumococci and influenza virus) are often synergistic, resulting in enhanced disease severity. Although colonization of the nasopharynx is the precursor to disease and transmission, little is known about bacterial-viral interactions that occur within this niche. In this study, we developed a novel mouse model to examine pneumococcal-viral interactions in the nasopharynx with pneumonia virus of mice (PVM) and influenza. We found that PVM infection benefits pneumococci by increasing their numbers in the nasopharynx and shedding of these bacteria in respiratory secretions. In contrast, we discovered that pneumococci decrease PVM numbers by accelerating viral clearance. We also report a similar effect of pneumococci on influenza. By showing that coinfections lead to both synergistic and antagonistic outcomes, our findings challenge the existing dogma in the field. Our work has important applications and implications for bacterial and viral vaccines that target these microbes.


Subject(s)
Antibiosis , Coinfection/microbiology , Coinfection/virology , Pneumococcal Infections/virology , Pneumovirus Infections/virology , Respiratory System/virology , Age Factors , Animals , Coinfection/immunology , Cytokines/analysis , Cytokines/immunology , Disease Models, Animal , Influenza A virus/genetics , Influenza A virus/immunology , Mice , Mice, Inbred C57BL , Murine pneumonia virus/genetics , Murine pneumonia virus/immunology , Nasopharynx/virology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Pneumovirus Infections/immunology , Respiratory System/immunology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/immunology , Viral Load
14.
Emerg Microbes Infect ; 11(1): 368-383, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34989330

ABSTRACT

Older individuals are at higher risk of SARS-CoV-2 infection and severe outcomes, but the underlying mechanisms are incompletely understood. In addition, how age modulates SARS-CoV-2 re-infection and vaccine breakthrough infections remain largely unexplored. Here, we investigated age-associated SARS-CoV-2 pathogenesis, immune responses, and the occurrence of re-infection and vaccine breakthrough infection utilizing a wild-type C57BL/6N mouse model. We demonstrated that interferon and adaptive antibody response upon SARS-CoV-2 challenge are significantly impaired in aged mice compared to young mice, which results in more effective virus replications and severe disease manifestations in the respiratory tract. Aged mice also showed increased susceptibility to re-infection due to insufficient immune protection acquired during the primary infection. Importantly, two-dose COVID-19 mRNA vaccination conferred limited adaptive immune response among the aged mice, making them susceptible to SARS-CoV-2 infection. Collectively, our findings call for tailored and optimized treatments and prevention strategies against SARS-CoV-2 among older individuals.


Subject(s)
Age Factors , COVID-19 Vaccines/immunology , COVID-19/immunology , SARS-CoV-2/immunology , Aging/immunology , Animals , Antibodies, Viral/immunology , COVID-19/pathology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/administration & dosage , Disease Models, Animal , Disease Susceptibility , Female , Humans , Immunity , Mice , Mice, Inbred C57BL , Respiratory System/immunology , Respiratory System/virology , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Vaccination , Virus Replication
15.
Sci Rep ; 12(1): 427, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013475

ABSTRACT

Neutrophilic airway inflammation is highly prevalent in racehorses in training, with the term mild to moderate equine asthma (MMEA) being applied to the majority of such cases. Our proposed study is largely derived from the strong association between MMEA in racehorses and their entry into a race training program. The objectives of this study are to characterise the effect of training on the local pulmonary immune system by defining the gene and protein expression of tracheal wash (TW) derived samples from Thoroughbred racehorses prior to and following commencement of race training. Multiomics analysis detected 2138 differentially expressed genes and 260 proteins during the training period. Gene and protein sets were enriched for biological processes related to acute phase response, oxidative stress, haemopoietic processes, as well as to immune response and inflammation. This study demonstrated TW samples to represent a rich source of airway cells, protein and RNA to study airway immunity in the horse and highlighted the benefits of a multiomics methodological approach to studying the dynamics of equine airway immunity. Findings likely reflect the known associations between race-training and both airway inflammation and bleeding, offering further insight into the potential mechanisms which underpin training associated airway inflammation.


Subject(s)
Horses/immunology , Physical Conditioning, Animal , Proteome , Respiratory System/immunology , Transcriptome , Animals , Gene Expression Profiling , Male , Respiratory System/cytology
16.
Regul Toxicol Pharmacol ; 129: 105112, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34973388

ABSTRACT

Some proteins, including enzymes, can induce allergic sensitization of various types, including allergic sensitization of the respiratory tract. There is now an increased understanding of the role that the skin plays in the development of IgE-mediated allergy and this prompts the question whether topical exposure to enzymes used widely in consumer cleaning products could result in allergic sensitization. Here, the evidence that proteins can interact with the skin immune system and the way they do so is reviewed, together with a consideration of the experience gained over decades of the use of enzymes in laundry and cleaning products. The conclusion drawn is that although transcutaneous sensitization to proteins can occur (typically through compromised skin) resulting in IgE antibody-mediated allergy, in practice such skin contact with enzymes used in laundry and cleaning products does not appear to pose a significant risk of allergic disease. Further, the evidence summarized in this publication support the view that proteins do not pose a risk of allergic contact dermatitis.


Subject(s)
Detergents/pharmacology , Enzymes/immunology , Hypersensitivity/etiology , Hypersensitivity/immunology , Skin/immunology , Allergens/immunology , Dermatitis, Allergic Contact/etiology , Dermatitis, Allergic Contact/immunology , Food Hypersensitivity/etiology , Food Hypersensitivity/immunology , Humans , Molecular Weight , Respiratory System/immunology
17.
Mucosal Immunol ; 15(2): 314-326, 2022 02.
Article in English | MEDLINE | ID: mdl-34686839

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) play an important role in allergic airway inflammation. Despite recent advances in defining molecular mechanisms that control ILC2 development and function, the role of endogenous metabolites in the regulation of ILC2s remains poorly understood. Herein, we demonstrated that bilirubin, an end product of heme catabolism, was a potent negative regulator of ILC2s. Bilirubin metabolism was found to be significantly induced during airway inflammation in mouse models. The administration of unconjugated bilirubin (UCB) dramatically suppressed ILC2 responses to interleukin (IL)-33 in mice, including cell proliferation and the production of effector cytokines. Furthermore, UCB significantly alleviated ILC2-driven airway inflammation, which was aggravated upon clearance of endogenous UCB. Mechanistic studies showed that the effects of bilirubin on ILC2s were associated with downregulation of ERK phosphorylation and GATA3 expression. Clinically, newborns with hyperbilirubinemia displayed significantly lower levels of ILC2 with impaired function and suppressed ERK signaling. Together, these findings indicate that bilirubin serves as an endogenous suppressor of ILC2s and might have potential therapeutic value in the treatment of allergic airway inflammation.


Subject(s)
Bilirubin , Lymphocytes , Respiratory Hypersensitivity , Animals , Bilirubin/pharmacology , Cytokines/metabolism , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-33/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology , Mice , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Respiratory System/immunology , Respiratory System/metabolism
18.
Int Arch Allergy Immunol ; 183(2): 153-159, 2022.
Article in English | MEDLINE | ID: mdl-34551414

ABSTRACT

Mast cells (MCs) are involved in several biological processes, such as defense against pathogens, immunomodulation, tissue repair after injury, and angiogenesis. MCs have been shown to change from protective immune cells to potent pro-inflammatory cells, influencing the progression of many pathological conditions, including autoimmune diseases and cancers. The role of MCs in the pathogenesis of rhinopathies has often been underestimated, since previous studies have focused their attention on eosinophils and neutrophils, while MCs were considered involved exclusively in allergic rhinitis. However, recent nasal cytology findings have shown the involvement of MCs in several rhinopathies, such as NARMA, NARESMA, and CRSwNP. These recent evidences highlight the crucial role that MCs play in orchestrating the inflammation of the nasal mucosa, through complex biological mechanisms, not yet fully understood. In this context, a better understanding of these mechanisms is fundamental for practicing Precision Medicine, which requires careful population selection and stratification into subgroups based on the phenotype/endotype of the patients, in order to guarantee the patient a tailored therapy. Based on this background, further studies are needed to understand the pathophysiological mechanisms involving MCs and, consequently, to develop targeted therapies aimed to obtain a selective inhibition of tissue remodeling and preventing MC-mediated immune suppression.


Subject(s)
Disease Susceptibility , Mast Cells/immunology , Mast Cells/metabolism , Pharyngeal Diseases/etiology , Pharyngeal Diseases/metabolism , Animals , Biomarkers , Cell Plasticity , Disease Management , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Neovascularization, Physiologic , Pharyngeal Diseases/diagnosis , Pharyngeal Diseases/therapy , Phenotype , Respiratory Physiological Phenomena , Respiratory System/immunology , Respiratory System/metabolism , Respiratory System/pathology , Rhinitis/etiology , Rhinitis/metabolism , Rhinitis/pathology
19.
Auris Nasus Larynx ; 49(1): 1-10, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34304944

ABSTRACT

The mucosal immune system prevents microorganism invasion through mucosal surfaces and consists of inductive and effector sites. Nasopharynx-associated lymphoid tissue (NALT) functions as an inductive site, inducing mucosal immune responses in the upper respiratory tract. It follows that intranasal vaccines may prevent upper respiratory infections. To induce and enhance the immune response by administering inactivated antigens intranasally, mucosal adjuvants have been developed, including mutant cholera toxin and cationic cholesteryl pullulan nanogel, which do not accumulate in the central nervous system. Moreover, multivalent pneumococcal polysaccharide conjugate vaccines are used to prevent invasive pneumococcal infections and otitis media, although they only provide moderate protection against acute otitis media because non-vaccine serotypes of Streptococcus pneumoniae and Haemophilus influenzae also cause this infection. To address this problem, pneumococcal surface protein A of S. pneumoniae and P6 of H. influenzae are used as broad-spectrum vaccine antigens. Alternatively, phosphorylcholine (PC) is present in the cell walls of both gram-positive and gram-negative bacteria and induces immune responses through antigenic activity. The significant effects of PC as a mucosal vaccine have been demonstrated through intranasal and sublingual immunization in mice. Furthermore, intranasal administration of PC reverses increases in IgE levels and prevents allergic rhinitis. After immunization with pneumococcal polysaccharide conjugate vaccine, intranasal immunization with PC boosts immune responses to vaccine strains and to PC itself. Thus, PC may be useful as a mucosal vaccine to prevent upper respiratory infections and allergic rhinitis, and it could be used as a booster to the currently used pneumococcal vaccine as it protects against non-vaccine strains.


Subject(s)
Immunity, Mucosal , Phosphorylcholine/immunology , Respiratory System/immunology , Vaccines , Administration, Intranasal , Animals , Antigens, Bacterial , Haemophilus influenzae/immunology , Humans , Immune System , Immunoglobulin A, Secretory , Mice , Mucous Membrane , Phosphorylcholine/therapeutic use , Pneumococcal Vaccines , Rhinitis, Allergic/prevention & control , Streptococcus pneumoniae/immunology , Vaccines/immunology
20.
J Leukoc Biol ; 111(1): 209-221, 2022 01.
Article in English | MEDLINE | ID: mdl-33857344

ABSTRACT

Communication between the nervous and immune systems serves a key role in host-protective immunity at mucosal barrier sites including the respiratory tract. In these tissues, neuroimmune interactions operate in bidirectional circuits that can sense and respond to mechanical, chemical, and biologic stimuli. Allergen- or helminth-induced products can produce airway inflammation by direct action on nociceptive afferents and adjacent tissues. The activity of nociceptive afferents can regulate innate and adaptive immune responses via neuropeptides and neurotransmitter signaling. This review will summarize recent work investigating the role of neuropeptides CGRP, VIP, neuromedins, substance P, and neurotransmitters dopamine and the B2-adrenoceptor agonists epinepherine/norepinepherine, each of which influence type 2 immunity by instructing mast cell, innate lymphoid cell type 2, dendritic cell, and T cell responses, both in the airway and the draining lymph node. Afferents in the airway also contain receptors for alarmins and cytokines, allowing their activity to be modulated by immune cell secreted products, particularly those secreted by mast cells. Taken together, we propose that further investigation of how immunoregulatory neuropeptides shape respiratory inflammation in experimental systems may reveal novel therapeutic targets for addressing the increasing prevalence of chronic airway disease in humans.


Subject(s)
Hypersensitivity/immunology , Inflammation/immunology , Neuroimmunomodulation , Neuropeptides/immunology , Animals , Humans , Immunity, Innate , Mucous Membrane/immunology , Respiratory System/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...