Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.903
Filter
1.
Exerc Immunol Rev ; 30: 6-13, 2024.
Article in English | MEDLINE | ID: mdl-39094181

ABSTRACT

Background: Several studies have reported that marathon runners have a higher risk of upper respiratory tract infections (URTI) post marathon than non-exercising controls. However, other studies did not find a higher risk of URTI in the same participants before and after a marathon, precluding a conclusive consensus. Besides the between-subjects effects, another important confounding factor in these results is the different pre and post follow-up time to track URTI. Objectives: Identify by meta-analysis whether a marathon Running increases the risk of URTI, adjusting the follow-up time to track URTI. Data sources: We searched for articles using MEDLINE (PubMed), Embase, Scopus, Web of Science, the Cochrane Library, and EBSCOhost, combining the marathon and respiratory infection descriptor synonyms, on 1st December 2022. Eligibility criteria: The PICOS framework included human population, comparison between pre and post marathon running, of URTI symptoms (assessed from one to 4 weeks), in noncontrolled intervention studies. Data Synthesis: Because follow-up was longer before the marathon in many studies, we adjusted the number of subjects with infections before marathon to the equivalent post-marathon follow-up duration. There was 18% higher incidence of URTI post-marathon (OR 1.18 95%CI [1.05-1.33], p= 0.005) in a very consistent meta-analysis (I2 = 0%, p = 0.69), with no risk of publication bias (Egger test p-value = 0.82) for the 7 studies included. The main issues with quality of the studies were bias in measuring the outcome, bias in classification of intervention (participation in the marathon) and time-varying confounding (corrected for analysis), and therefore the quality of evidence was moderate (GRADE approach = 3). Limitations: The need for follow-up time adjustment is a limitation, since the number of URTI recorded could be different if the original studies had used the same follow-up time pre and post marathon. The subjectivity of the URTI assessments is another limitation in this field. Conclusions: There is an increased risk of URTI post marathon running and research on this topic to understand mechanisms might support runners to find efficient interventions to reduce this risk. Protocol: Protocol registration on in the International Prospective Register of Systematic Reviews (PROSPERO): CRD42022380991.


Subject(s)
Marathon Running , Respiratory Tract Infections , Humans , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/immunology , Risk Factors , Exercise/physiology , Running/physiology , Immune System , Risk
2.
Eur Respir Rev ; 33(172)2024 Apr.
Article in English | MEDLINE | ID: mdl-39009407

ABSTRACT

Respiratory viral infections frequently lead to severe respiratory disease, particularly in vulnerable populations such as young children, individuals with chronic lung conditions and older adults, resulting in hospitalisation and, in some cases, fatalities. The innate immune system plays a crucial role in monitoring for, and initiating responses to, viruses, maintaining a state of preparedness through the constant expression of antimicrobial defence molecules. Throughout the course of infection, innate immunity remains actively involved, contributing to viral clearance and damage control, with pivotal contributions from airway epithelial cells and resident and newly recruited immune cells. In instances where viral infections persist or are not effectively eliminated, innate immune components prominently contribute to the resulting pathophysiological consequences. Even though both young children and older adults are susceptible to severe respiratory disease caused by various respiratory viruses, the underlying mechanisms may differ significantly. Children face the challenge of developing and maturing their immunity, while older adults contend with issues such as immune senescence and inflammaging. This review aims to compare the innate immune responses in respiratory viral infections across both age groups, identifying common central hubs that could serve as promising targets for innovative therapeutic and preventive strategies, despite the apparent differences in underlying mechanisms.


Subject(s)
Host-Pathogen Interactions , Immunity, Innate , Respiratory Tract Infections , Virus Diseases , Humans , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Respiratory Tract Infections/epidemiology , Age Factors , Virus Diseases/immunology , Virus Diseases/epidemiology , Aged , Child , Adult , Adolescent , Child, Preschool , Middle Aged , Immunosenescence/immunology , Young Adult , Lung/immunology , Lung/virology , Infant , Animals , Signal Transduction
3.
Respir Res ; 25(1): 277, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39010108

ABSTRACT

To protect against the constant threat of inhaled pathogens, the lung is equipped with cellular defenders. In coordination with resident and recruited immune cells, this defence is initiated by the airway and alveolar epithelium following their infection with respiratory viruses. Further support for viral clearance and infection resolution is provided by adjacent endothelial and stromal cells. However, even with these defence mechanisms, respiratory viral infections are a significant global health concern, causing substantial morbidity, socioeconomic losses, and mortality, underlining the need to develop effective vaccines and antiviral medications. In turn, the identification of new treatment options for respiratory infections is critically dependent on the availability of tractable in vitro experimental models that faithfully recapitulate key aspects of lung physiology. For such models to be informative, it is important these models incorporate human-derived, physiologically relevant versions of all cell types that normally form part of the lungs anti-viral response. This review proposes a guideline using human induced pluripotent stem cells (iPSCs) to create all the disease-relevant cell types. iPSCs can be differentiated into lung epithelium, innate immune cells, endothelial cells, and fibroblasts at a large scale, recapitulating in vivo functions and providing genetic tractability. We advocate for building comprehensive iPSC-derived in vitro models of both proximal and distal lung regions to better understand and model respiratory infections, including interactions with chronic lung diseases.


Subject(s)
Induced Pluripotent Stem Cells , Lung , Respiratory Tract Infections , Virus Diseases , Humans , Lung/immunology , Lung/virology , Respiratory Tract Infections/virology , Respiratory Tract Infections/immunology , Virus Diseases/immunology , Animals , Cell Differentiation/physiology , Models, Biological
4.
Clin Epigenetics ; 16(1): 85, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961479

ABSTRACT

BACKGROUND: Infants with frequent viral and bacterial respiratory infections exhibit compromised immunity to routine immunizations. They are also more likely to develop chronic respiratory diseases in later childhood. This study investigated the feasibility of epigenetic profiling to reveal endotype-specific molecular pathways with potential for early identification and immuno-modulation. Peripheral blood mononuclear cells from respiratory infection allergy/asthma-prone (IAP) infants and non-infection allergy/asthma prone (NIAP) were retrospectively selected for genome-wide DNA methylation and single nucleotide polymorphism analysis. The IAP infants were enriched for the low vaccine responsiveness (LVR) phenotype (Fisher's exact p-value = 0.02). RESULTS: An endotype signature of 813 differentially methylated regions (DMRs) comprising 238 lead CpG associations (FDR < 0.05) emerged, implicating pathways related to asthma, mucin production, antigen presentation and inflammasome activation. Allelic variation explained only a minor portion of this signature. Stimulation of mononuclear cells with monophosphoryl lipid A (MPL), a TLR agonist, partially reversed this signature at a subset of CpGs, suggesting the potential for epigenetic remodeling. CONCLUSIONS: This proof-of-concept study establishes a foundation for precision endotyping of IAP children and highlights the potential for immune modulation strategies using adjuvants for future investigation.


Subject(s)
Asthma , DNA Methylation , Epigenesis, Genetic , Leukocytes, Mononuclear , Respiratory Tract Infections , Humans , Asthma/genetics , Asthma/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , DNA Methylation/genetics , Male , Female , Respiratory Tract Infections/immunology , Respiratory Tract Infections/genetics , Infant , Epigenesis, Genetic/genetics , Polymorphism, Single Nucleotide , CpG Islands/genetics , Retrospective Studies , Genome-Wide Association Study/methods , Child, Preschool , Child , Proof of Concept Study
5.
Hum Vaccin Immunother ; 20(1): 2368288, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38953250

ABSTRACT

Many pathogens enter the host through mucosal sites. Thus, interfering with pathogen entry through local neutralization at mucosal sites therefore is an effective strategy for preventing disease. Mucosally administered vaccines have the potential to induce protective immune responses at mucosal sites. This manuscript delves into some of the latest developments in mucosal vaccination, particularly focusing on advancements in adjuvant technologies and the role of these adjuvants in enhancing vaccine efficacy against respiratory pathogens. It highlights the anatomical and immunological complexities of the respiratory mucosal immune system, emphasizing the significance of mucosal secretory IgA and tissue-resident memory T cells in local immune responses. We further discuss the differences between immune responses induced through traditional parenteral vaccination approaches vs. mucosal administration strategies, and explore the protective advantages offered by immunization through mucosal routes.


Subject(s)
Immunity, Mucosal , Respiratory Mucosa , Humans , Respiratory Mucosa/immunology , Animals , Vaccines/immunology , Vaccines/administration & dosage , Administration, Mucosal , Adjuvants, Vaccine , Vaccination/methods , Adjuvants, Immunologic/administration & dosage , Respiratory Tract Infections/immunology , Respiratory Tract Infections/prevention & control , Memory T Cells/immunology , Immunoglobulin A, Secretory/immunology
6.
Eur Respir Rev ; 33(173)2024 Jul.
Article in English | MEDLINE | ID: mdl-38960615

ABSTRACT

Bronchiectasis is marked by bronchial dilatation, recurrent infections and significant morbidity, underpinned by a complex interplay between microbial dysbiosis and immune dysregulation. The identification of distinct endophenotypes have refined our understanding of its pathogenesis, including its heterogeneous disease mechanisms that influence treatment and prognosis responses. Next-generation sequencing (NGS) has revolutionised the way we view airway microbiology, allowing insights into the "unculturable". Understanding the bronchiectasis microbiome through targeted amplicon sequencing and/or shotgun metagenomics has provided key information on the interplay of the microbiome and host immunity, a central feature of disease progression. The rapid increase in translational and clinical studies in bronchiectasis now provides scope for the application of precision medicine and a better understanding of the efficacy of interventions aimed at restoring microbial balance and/or modulating immune responses. Holistic integration of these insights is driving an evolving paradigm shift in our understanding of bronchiectasis, which includes the critical role of the microbiome and its unique interplay with clinical, inflammatory, immunological and metabolic factors. Here, we review the current state of infection and the microbiome in bronchiectasis and provide views on the future directions in this field.


Subject(s)
Bronchiectasis , Dysbiosis , Host-Pathogen Interactions , Microbiota , Bronchiectasis/microbiology , Bronchiectasis/immunology , Humans , Lung/microbiology , Animals , Risk Factors , Bacteria/genetics , Bacteria/classification , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/immunology , Prognosis
7.
Front Immunol ; 15: 1389920, 2024.
Article in English | MEDLINE | ID: mdl-38957464

ABSTRACT

Probiotic consumption strongly influences local intestinal immunity and systemic immune status. Heyndrickxia coagulans strain SANK70258 (HC) is a spore-forming lactic acid bacterium that has immunostimulatory properties on peripheral tissues. However, few reports have examined the detailed effectiveness of HC on human immune function and its mechanism of action. Therefore, we conducted a randomized, double-blind, placebo-controlled, parallel-group study to comprehensively evaluate the effects of HC on immunostimulatory capacity, upper respiratory tract infection (URTI) symptoms, and changes in intestinal organic-acid composition. Results of a questionnaire survey of URTI symptoms showed that runny nose, nasal congestion, sneezing, and sore throat scores as well as the cumulative number of days of these symptoms were significantly lower in the HC group than in the placebo group during the study period. Furthermore, the salivary secretory immunoglobulin A (sIgA) concentration was significantly higher, and the natural killer (NK) cell activity tended to be higher in the HC group than in the placebo group. In addition, we performed an exposure culture assay of inactivated influenza virus on peripheral blood mononuclear cells (PBMCs) isolated from the blood of participants in the HC and placebo groups. Gene-expression analysis in PBMCs after culture completion showed that IFNα and TLR7 expression levels were significantly higher in the HC group than in the placebo group. In addition, the expression levels of CD304 tended to be higher in the HC group than in the placebo group. On the other hand, the HC group showed a significantly higher increase in the intestinal butyrate concentration than the placebo group. HC intake also significantly suppressed levels of IL-6 and TNFα produced by PBMCs after exposure to inactivated influenza virus. Collectively, these results suggest that HC activated plasmacytoid dendritic cells expressing TLR7 and CD304 and strongly induced IFNα production, subsequently activating NK cells and increasing sIgA levels, and induced anti-inflammatory effects via increased intestinal butyrate levels. These changes may contribute to the acquisition of host resistance to viral infection and URTI prevention.


Subject(s)
Probiotics , Respiratory Tract Infections , Humans , Respiratory Tract Infections/immunology , Double-Blind Method , Male , Adult , Probiotics/administration & dosage , Female , Young Adult , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Gastrointestinal Microbiome/immunology , Immunoglobulin A, Secretory/immunology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/immunology , Immunomodulation
8.
Int J Mol Sci ; 25(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38892370

ABSTRACT

The respiratory system is constantly exposed to viral infections that are responsible for mild to severe diseases. In this narrative review, we focalized the attention on respiratory syncytial virus (RSV), influenza virus, and severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infections, responsible for high morbidity and mortality in the last decades. We reviewed the human innate and adaptive immune responses in the airways following infection, focusing on a particular population: newborns and pregnant women. The recent Coronavirus disease-2019 (COVID-19) pandemic has highlighted how our interest in viral pathologies must not decrease. Furthermore, we must increase our knowledge of infection mechanisms to improve our future defense strategies.


Subject(s)
COVID-19 , Respiratory Tract Infections , SARS-CoV-2 , Humans , COVID-19/immunology , COVID-19/virology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Pregnancy , SARS-CoV-2/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Female , Immunity, Innate , Adaptive Immunity , Infant, Newborn , Influenza, Human/immunology , Influenza, Human/virology , Virus Diseases/immunology
9.
Front Immunol ; 15: 1371118, 2024.
Article in English | MEDLINE | ID: mdl-38873612

ABSTRACT

Background: The respiratory tract microbiome is essential for human health and well-being and is determined by genetic, lifestyle, and environmental factors. Patients with Common Variable Immunodeficiency (CVID) suffer from respiratory and intestinal tract infections, leading to chronic diseases and increased mortality rates. While CVID patients' gut microbiota have been analyzed, data on the respiratory microbiome ecosystem are limited. Objective: This study aims to analyze the bacterial composition of the oropharynx of adults with CVID and its link with clinical and immunological features and risk for respiratory acute infections. Methods: Oropharyngeal samples from 72 CVID adults and 26 controls were collected in a 12-month prospective study. The samples were analyzed by metagenomic bacterial 16S ribosomal RNA sequencing and processed using the Quantitative Insights Into Microbial Ecology (QIME) pipeline. Differentially abundant species were identified and used to build a dysbiosis index. A machine learning model trained on microbial abundance data was used to test the power of microbiome alterations to distinguish between healthy individuals and CVID patients. Results: Compared to controls, the oropharyngeal microbiome of CVID patients showed lower alpha- and beta-diversity, with a relatively increased abundance of the order Lactobacillales, including the family Streptococcaceae. Intra-CVID analysis identified age >45 years, COPD, lack of IgA, and low residual IgM as associated with a reduced alpha diversity. Expansion of Haemophilus and Streptococcus genera was observed in patients with undetectable IgA and COPD, independent from recent antibiotic use. Patients receiving azithromycin as antibiotic prophylaxis had a higher dysbiosis score. Expansion of Haemophilus and Anoxybacillus was associated with acute respiratory infections within six months. Conclusions: CVID patients showed a perturbed oropharynx microbiota enriched with potentially pathogenic bacteria and decreased protective species. Low residual levels of IgA/IgM, chronic lung damage, anti antibiotic prophylaxis contributed to respiratory dysbiosis.


Subject(s)
Common Variable Immunodeficiency , Dysbiosis , Oropharynx , Respiratory Tract Infections , Humans , Common Variable Immunodeficiency/microbiology , Common Variable Immunodeficiency/immunology , Common Variable Immunodeficiency/complications , Oropharynx/microbiology , Male , Female , Middle Aged , Adult , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/immunology , Microbiota , Prospective Studies , Aged , RNA, Ribosomal, 16S/genetics , Acute Disease , Bacteria/classification , Bacteria/genetics , Case-Control Studies
10.
Cell Immunol ; 401-402: 104841, 2024.
Article in English | MEDLINE | ID: mdl-38878619

ABSTRACT

Pneumonia persists as a public health crisis, representing the leading cause of death due to infection. Whether respiratory tract infections progress to pneumonia and its sequelae such as acute respiratory distress syndrome and sepsis depends on numerous underlying conditions related to both the causative agent and host. Regarding the former, pneumonia burden remains staggeringly high, despite the effectiveness of pathogen-targeting strategies such as vaccines and antibiotics. This demands a greater understanding of host features that collaborate to promote immune resistance and tissue resilience in the infected lung. Such features inside the pulmonary compartment have drawn much attention, where major advances have been made related to resident and recruited immune activity. By comparison, extra-pulmonary processes guiding pneumonia susceptibility are relatively elusive, constituting the focus of this review. Here we will highlight examples of when, how, and why tissues outside of the lungs dispatch signals that modulate local immunity in the airspaces. Topics include the liver, gut, bone marrow, brain and more, all of which contribute in direct and indirect ways to pneumonia outcome. When tuned appropriately, it has become clear that these responses can serve protective roles, and this will be considered distinctly from what would otherwise be aberrant responses characteristic of pneumonia-induced organ injury and sepsis. Further advances in this area may reveal novel targetable areas for clinical intervention that are not confined to the intra-pulmonary space.


Subject(s)
Lung , Humans , Animals , Lung/immunology , Pneumonia/immunology , Liver/immunology , Respiratory Tract Infections/immunology , Brain/immunology , Sepsis/immunology
11.
Toxicol Appl Pharmacol ; 489: 117010, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38901696

ABSTRACT

Humoral responses to respiratory viruses, such as influenza viruses, develop over time and are central to protection from repeated infection with the same or similar viruses. Epidemiological and experimental studies have linked exposures to environmental contaminants that bind the aryl hydrocarbon receptor (AHR) with modulated antibody responses to pathogenic microorganisms and common vaccinations. Other studies have prompted investigation into the potential therapeutic applications of compounds that activate AHR. Herein, using two different AHR ligands [2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and 2-(1H-Indol-3-ylcarbonyl)-4-thiazolecarboxylic acid methyl ester (ITE), to modulate the duration of AHR activity, we show that the humoral response to viral infection is dependent upon the duration and timing of AHR signaling, and that different cellular elements of the response have different sensitivities. When AHR activation was initiated prior to infection with influenza A virus, there was suppression of all measured elements of the humoral response (i.e., the frequency of T follicular helper cells, germinal center B cells, plasma cells, and circulating virus-specific antibody). However, when the timing of AHR activation was adjusted to either early (days -1 to +5 relative to infection) or later (days +5 onwards), then AHR activation affected different aspects of the overall humoral response. These findings highlight the importance of considering the timing of AHR activation in relation to triggering an immune response, particularly when targeting the AHR to manipulate disease processes.


Subject(s)
Immunity, Humoral , Polychlorinated Dibenzodioxins , Receptors, Aryl Hydrocarbon , Receptors, Aryl Hydrocarbon/metabolism , Animals , Immunity, Humoral/drug effects , Polychlorinated Dibenzodioxins/toxicity , Female , Time Factors , Mice , Mice, Inbred C57BL , Indoles/pharmacology , Signal Transduction/drug effects , Antibodies, Viral , Orthomyxoviridae Infections/immunology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Respiratory Tract Infections/metabolism , Ligands , Thiazoles
12.
Biol Open ; 13(6)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38875000

ABSTRACT

Viral infectivity depends on multiple factors. Recent studies showed that the interaction between viral RNAs and endogenous microRNAs (miRNAs) regulates viral infectivity; viral RNAs function as a sponge of endogenous miRNAs and result in upregulation of its original target genes, while endogenous miRNAs target viral RNAs directly and result in repression of viral gene expression. In this study, we analyzed the possible interaction between parainfluenza virus RNA and endogenous miRNAs in human and mouse lungs. We showed that the parainfluenza virus can form base pairs with human miRNAs abundantly than mouse miRNAs. Furthermore, we analyzed that the sponge effect of endogenous miRNAs on viral RNAs may induce the upregulation of transcription regulatory factors. Then, we performed RNA-sequence analysis and observed the upregulation of transcription regulatory factors in the early stages of parainfluenza virus infection. Our studies showed how the differential expression of endogenous miRNAs in lungs could contribute to respiratory virus infection and species- or tissue-specific mechanisms and common mechanisms could be conserved in humans and mice and regulated by miRNAs during viral infection.


Subject(s)
Lung , MicroRNAs , Animals , MicroRNAs/genetics , Mice , Humans , Lung/virology , Lung/immunology , Lung/metabolism , RNA, Viral/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Gene Expression Regulation , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Respiratory Tract Infections/genetics , Respirovirus Infections/immunology
13.
Pediatr Allergy Immunol ; 35(6): e14184, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38924159

ABSTRACT

Asthma is the most common chronic disease in childhood affecting the daily lives of many patients despite current treatment regimens. Therefore, the need for new therapeutic approaches is evident, where a primary prevention strategy is the ultimate goal. Studies of children born to mothers in farming environments have shown a lower risk of respiratory infections and asthma development. Already at birth, these newborns have demonstrated accelerated maturation and upregulation of host defense immune functions suggesting a prenatal transplacental training of the innate immune system through maternal microbial exposure. This mechanism could possibly be utilized to help prevent both respiratory infections and asthma in young children. Human studies exploring the potential preventative effects of pregnancy bacterial lysate treatment on asthma and respiratory infections are lacking, however, this has been studied in experimental studies using mice through administrations of the bacterial lysate OM-85. This review will present the current literature on the immunomodulatory effects relevant for respiratory infections and asthma in the offspring of mice treated with OM-85 throughout pregnancy. Further, the review will discuss the cellular and molecular mechanisms behind these effects. In conclusion, we found promising results of an accelerated immune competence and improved resistance to airway challenges as a result of prenatal bacterial lysate treatment that may pave the way for implementing this in human trials to prevent asthma and respiratory infections.


Subject(s)
Asthma , Disease Models, Animal , Prenatal Exposure Delayed Effects , Respiratory Tract Infections , Animals , Asthma/prevention & control , Asthma/immunology , Pregnancy , Female , Humans , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/immunology , Mice , Prenatal Exposure Delayed Effects/immunology , Cell Extracts/therapeutic use , Bacterial Lysates
14.
Viruses ; 16(6)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38932273

ABSTRACT

The epidemiology of different respiratory viral infections is believed to be affected by prior viral infections in addition to seasonal effects. This PROSPERO-registered systematic review identified 7388 studies, of which six met our criteria to answer the question specifically. The purpose of this review was to compare the prevalence of sequential viral infections in those with previously documented positive versus negative swabs. The pooled prevalence of sequential viral infections over varying periods from 30-1000 days of follow-up was higher following a negative respiratory viral swab at 0.15 than following a positive swab at 0.08, indicating the potential protective effects of prior respiratory viral infections. However, significant heterogeneity and publication biases were noted. There is some evidence, albeit of low quality, of a possible protective effect of an initial viral infection against subsequent infections by a different virus, which is possibly due to broad, nonspecific innate immunity. Future prospective studies are needed to validate our findings.


Subject(s)
Cross Protection , Respiratory Tract Infections , Virus Diseases , Humans , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/prevention & control , Virus Diseases/immunology , Virus Diseases/prevention & control , Cross Protection/immunology , Prevalence
15.
Eur Respir Rev ; 33(172)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38811032

ABSTRACT

Respiratory viral infections represent one of the major causes of death worldwide. The recent coronavirus disease 2019 pandemic alone claimed the lives of over 6 million people around the globe. It is therefore crucial to understand how the immune system responds to these threats and how respiratory infection can be controlled and constrained. Dendritic cells (DCs) are one of the key players in antiviral immunity because of their ability to detect pathogens. They can orchestrate an immune response that will, in most cases, lead to viral clearance. Different subsets of DCs are present in the lung and each subset can contribute to antiviral responses through various mechanisms. In this review, we discuss the role of the different lung DC subsets in response to common respiratory viruses, with a focus on respiratory syncytial virus, influenza A virus and severe acute respiratory syndrome coronavirus 2. We also review how lung DC-mediated responses to respiratory viruses can lead to the worsening of an existing chronic pulmonary disease such as asthma. Throughout the review, we discuss results obtained from animal studies as well as results generated from infected patients.


Subject(s)
Dendritic Cells , Respiratory Tract Infections , Dendritic Cells/immunology , Dendritic Cells/virology , Humans , Animals , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Lung/immunology , Lung/virology , Host-Pathogen Interactions , COVID-19/immunology , Virus Diseases/immunology , Virus Diseases/virology , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity
16.
Viruses ; 16(5)2024 05 06.
Article in English | MEDLINE | ID: mdl-38793615

ABSTRACT

Reflecting on this Special Issue dedicated to pediatric respiratory viruses, it is evident that the shadow cast by the global SARS-CoV-2 pandemic has profoundly impacted individuals of all ages and backgrounds, neonates and school-aged children being vulnerable cohorts resulting from the evolving immunological profiles and limited exposures to immunity-building experienced during this unprecedented era [...].


Subject(s)
COVID-19 , Respiratory Tract Infections , SARS-CoV-2 , Humans , Child , COVID-19/immunology , COVID-19/virology , Respiratory Tract Infections/virology , Respiratory Tract Infections/immunology , SARS-CoV-2/immunology , Child, Preschool , Infant, Newborn , Infant , Virus Diseases/immunology , Virus Diseases/virology
17.
Curr Opin Virol ; 66: 101410, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718575

ABSTRACT

Viral infections, including those affecting the respiratory tract, can alter the composition of the intestinal microbiota, which, in turn, can significantly influence both innate and adaptive immune responses, resulting in either enhanced pathogen clearance or exacerbation of the infection, possibly leading to inflammatory complications. A deeper understanding of the interplay between the intestinal microbiota and host immune responses in the context of respiratory viral infections (i.e. the gut-lung axis) is necessary to develop new treatments. This review highlights key mechanisms by which the intestinal microbiota, including its metabolites, can act locally or at distant organs to combat respiratory viruses. Therapeutics aimed at harnessing the microbiota to prevent and/or help treat respiratory viral infections represent a promising avenue for future investigation.


Subject(s)
Gastrointestinal Microbiome , Immunity, Innate , Respiratory Tract Infections , Virus Diseases , Humans , Gastrointestinal Microbiome/immunology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Virus Diseases/immunology , Virus Diseases/virology , Animals , Adaptive Immunity , Lung/microbiology , Lung/immunology , Lung/virology
18.
Hum Cell ; 37(4): 1080-1090, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38814518

ABSTRACT

Airway epithelium represents a physical barrier against toxic substances and pathogens but also presents pattern recognition receptors on the epithelial cells that detect pathogens leading to molecule release and sending signals that activate both the innate and adaptive immune responses. Thus, impaired airway epithelial function and poor integrity may increase the recurrence of infections. Probiotic use in respiratory diseases as adjuvant of traditional therapy is increasingly widespread. There is growing interest in the use of non-viable heat-killed bacteria, such as tyndallized bacteria (TB), due to safety concerns and to their immunomodulatory properties. This study explores in vitro the effects of a TB blend on the immune activation of airway epithelium. 16HBE bronchial epithelial cells were exposed to different concentrations of TB. Cell viability, TB internalization, TLR2 expression, IL-6, IL-8 and TGF-ßl expression/release, E-cadherin expression and wound healing were assessed. We found that TB were tolerated, internalized, increased TLR2, E-cadherin expression, IL-6 release and wound healing but decreased both IL-8 and TGF-ßl release. In conclusion, TB activate TLR2 pathway without inducing a relevant pro-inflammatory response and improve barrier function, leading to the concept that TB preserve epithelial homeostasis and could be used as strategy to prevent and to manage respiratory infection, exacerbations included.


Subject(s)
Bronchi , Epithelial Cells , Immunity, Innate , Toll-Like Receptor 2 , Humans , Toll-Like Receptor 2/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Bronchi/cytology , Bronchi/immunology , Interleukin-6/metabolism , Probiotics , Respiratory Mucosa/immunology , Cadherins/metabolism , Gene Expression , Cells, Cultured , Interleukin-8/metabolism , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Cell Survival
19.
Pathog Dis ; 822024 Feb 07.
Article in English | MEDLINE | ID: mdl-38714349

ABSTRACT

Respiratory pathogens can cause severe disease and even death, especially in the very young and very old. Studies investigating their prevalence often focus on individuals presenting to healthcare providers with symptoms. However, the design of prevention strategies, e.g. which target groups to vaccinate, will benefit from knowledge on the prevalence of, risk factors for and host response to these pathogens in the general population. In this study, upper respiratory samples (n = 1311) were collected cross-sectionally during winter from 11- and 24-month old children, their parents, and adults ≥60 years of age that were recruited irrespective of seeking medical care. Almost all children, approximately two-thirds of parents and a quarter of older adults tested positive for at least one pathogen, often in the absence of symptoms. Viral interference was evident for the combination of rhinovirus and respiratory syncytial virus. Attending childcare facilities and having siblings associated with increased pathogen counts in children. On average, children showed increased levels of mucosal cytokines compared to parents and especially proinflammatory molecules associated with the presence of symptoms. These findings may guide further research into transmission patterns of respiratory pathogens and assist in determining the most appropriate strategies for the prediction and prevention of disease.


Subject(s)
Cytokines , Respiratory Tract Infections , Seasons , Humans , Cross-Sectional Studies , Netherlands/epidemiology , Infant , Male , Female , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/virology , Respiratory Tract Infections/immunology , Prevalence , Middle Aged , Adult , Cytokines/metabolism , Aged , Child, Preschool , Aged, 80 and over , Virus Diseases/epidemiology , Virus Diseases/virology , Virus Diseases/immunology , Viruses/isolation & purification , Viruses/classification , Viruses/immunology
20.
J Virol ; 98(6): e0160423, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38780249

ABSTRACT

The global burden of disease caused by influenza B virus (IBV) is substantial; however, IBVs remain overlooked. Understanding host-pathogen interactions and establishing physiologically relevant models of infection are important for the development and assessment of therapeutics and vaccines against IBV. In this study, we assessed an upper respiratory tract (URT)-restricted model of mouse IBV infection, comparing it to the conventional administration of the virus to the total respiratory tract (TRT). We found that URT infections caused by different strains of IBV disseminate to the trachea but resulted in limited dissemination of IBV to the lungs. Infection of the URT did not result in weight loss or systemic inflammation even at high inoculum doses and despite robust viral replication in the nose. Dissemination of IBV to the lungs was enhanced in mice lacking functional type I IFN receptor (IFNAR2), but not IFNγ. Conversely, in mice expressing the IFN-inducible gene Mx1, we found reduced IBV replication in the lungs and reduced dissemination of IBV from the URT to the lungs. Inoculation of IBV in both the URT and TRT resulted in seroconversion against IBV. However, priming at the TRT conferred superior protection from a heterologous lethal IBV challenge compared to URT priming, as determined by improved survival rates and reduced viral replication throughout the respiratory tract. Overall, our study establishes a URT-restricted IBV infection model, highlights the critical role of IFNs in limiting dissemination of IBV to the lungs, and also demonstrates that the lack of viral replication in the lungs may impact protection from subsequent infections. IMPORTANCE: Our study investigated how influenza B virus (IBV) spreads from the nose to the lungs of mice and the impact this has on disease and protection from re-infection. We found that when applied to the nose only, IBV does not spread very efficiently to the lungs in a process controlled by the interferon response. Priming immunity at the nose only resulted in less protection from re-infection than priming immunity at both the nose and lungs. These insights can guide the development of potential therapies targeting the interferon response as well as of intranasal vaccines against IBV.


Subject(s)
Influenza B virus , Lung , Orthomyxoviridae Infections , Virus Replication , Animals , Mice , Influenza B virus/physiology , Influenza B virus/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Lung/virology , Lung/immunology , Disease Models, Animal , Interferons/metabolism , Interferons/immunology , Myxovirus Resistance Proteins/metabolism , Myxovirus Resistance Proteins/genetics , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/deficiency , Mice, Inbred C57BL , Host-Pathogen Interactions/immunology , Respiratory Tract Infections/virology , Respiratory Tract Infections/immunology , Female , Interferon-gamma/metabolism , Trachea/virology
SELECTION OF CITATIONS
SEARCH DETAIL