Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 852
Filter
1.
J Microbiol ; 62(9): 709-725, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39240507

ABSTRACT

The importance of the complex interplay between the microbiome and mucosal immunity, particularly within the respiratory tract, has gained significant attention due to its potential implications for the severity and progression of lung diseases. Therefore, this review summarizes the specific interactions through which the respiratory tract-specific microbiome influences mucosal immunity and ultimately impacts respiratory health. Furthermore, we discuss how the microbiome affects mucosal immunity, considering tissue-specific variations, and its capacity in respiratory diseases containing asthma, chronic obstructive pulmonary disease, and lung cancer. Additionally, we investigate the external factors which affect the relationship between respiratory microbiome and mucosal immune responses. By exploring these intricate interactions, this review provides valuable insights into the potential for microbiome-based interventions to modulate mucosal immunity and alleviate the severity of respiratory diseases.


Subject(s)
Disease Progression , Immunity, Mucosal , Microbiota , Humans , Microbiota/immunology , Asthma/immunology , Asthma/microbiology , Respiratory System/microbiology , Respiratory System/immunology , Animals , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/microbiology , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/microbiology , Lung Neoplasms/immunology , Lung Neoplasms/microbiology
2.
Front Cell Infect Microbiol ; 14: 1388222, 2024.
Article in English | MEDLINE | ID: mdl-38988815

ABSTRACT

Intramuscular vaccines present limitations in eliciting robust mucosal immunity and preventing respiratory pathogens transmission. Sublingual vaccine administration offers promising advantages, including interconnected mucosal protection. Despite these advantages, only a few clinical trials have explored sublingual vaccines, underscoring the necessity of optimizing next-generation vaccine formulas. Critical research priorities include understanding vector behavior in the oral environment, understanding their interactions with mucosal immunity and developing formulations enabling sustained mucosal contact to facilitate efficient transduction. Consequently, tonsil organoids, as representative human mucosal models, could offer critical insights into sublingual immunization. Thus, a multi-disciplinary approach integrating pharmacological, immunological, and manufacturing considerations is pivotal for sublingual vaccines in targeting pathogen-aggravated prevalent respiratory diseases including asthma, COPD and lung cancer, as well as the antimicrobial resistance crisis.


Subject(s)
Immunity, Mucosal , Vaccines , Humans , Vaccines/immunology , Vaccines/administration & dosage , Animals , Administration, Sublingual , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/prevention & control , Mouth/microbiology
3.
Eur Respir Rev ; 33(172)2024 Apr.
Article in English | MEDLINE | ID: mdl-38925790

ABSTRACT

Neuroimmune recognition and regulation in the respiratory system is a complex and highly coordinated process involving interactions between the nervous and immune systems to detect and respond to pathogens, pollutants and other potential hazards in the respiratory tract. This interaction helps maintain the health and integrity of the respiratory system. Therefore, understanding the complex interactions between the respiratory nervous system and immune system is critical to maintaining lung health and developing treatments for respiratory diseases. In this review, we summarise the projection distribution of different types of neurons (trigeminal nerve, glossopharyngeal nerve, vagus nerve, spinal dorsal root nerve, sympathetic nerve) in the respiratory tract. We also introduce several types of cells in the respiratory epithelium that closely interact with nerves (pulmonary neuroendocrine cells, brush cells, solitary chemosensory cells and tastebuds). These cells are primarily located at key positions in the respiratory tract, where nerves project to them, forming neuroepithelial recognition units, thus enhancing the ability of neural recognition. Furthermore, we summarise the roles played by these different neurons in sensing or responding to specific pathogens (influenza, severe acute respiratory syndrome coronavirus 2, respiratory syncytial virus, human metapneumovirus, herpes viruses, Sendai parainfluenza virus, Mycobacterium tuberculosis, Pseudomonas aeruginosa, Staphylococcus aureus, amoebae), allergens, atmospheric pollutants (smoking, exhaust pollution), and their potential roles in regulating interactions among different pathogens. We also summarise the prospects of bioelectronic medicine as a third therapeutic approach following drugs and surgery, as well as the potential mechanisms of meditation breathing as an adjunct therapy.


Subject(s)
Neuroimmunomodulation , Respiratory System , Humans , Animals , Respiratory System/immunology , Respiratory System/virology , Host-Pathogen Interactions , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/therapy , Respiratory Tract Diseases/physiopathology , Respiratory Tract Diseases/virology , Signal Transduction
4.
Nat Rev Microbiol ; 22(8): 492-506, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38778224

ABSTRACT

Communication between the gut and remote organs, such as the brain or the cardiovascular system, has been well established and recent studies provide evidence for a potential bidirectional gut-airway axis. Observations from animal and human studies indicate that respiratory insults influence the activity of the gut microbiome and that microbial ligands and metabolic products generated by the gut microbiome shape respiratory immunity. Information exchange between these two large mucosal surface areas regulates microorganism-immune interactions, with significant implications for the clinical and treatment outcomes of a range of respiratory conditions, including asthma, chronic obstructive pulmonary disease and lung cancer. In this Review, we summarize the most recent data in this field, offering insights into mechanisms of gut-airway crosstalk across spatial and temporal gradients and their relevance for respiratory health.


Subject(s)
Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/physiology , Animals , Respiratory Tract Diseases/microbiology , Respiratory Tract Diseases/immunology , Respiratory System/microbiology , Respiratory System/immunology , Asthma/microbiology , Asthma/immunology
5.
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
6.
J Allergy Clin Immunol ; 153(6): 1647-1654, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38309597

ABSTRACT

BACKGROUND: Farm exposures in early life reduce the risks for childhood allergic diseases and asthma. There is less information about how farm exposures relate to respiratory illnesses and mucosal immune development. OBJECTIVE: We hypothesized that children raised in farm environments have a lower incidence of respiratory illnesses over the first 2 years of life than nonfarm children. We also analyzed whether farm exposures or respiratory illnesses were related to patterns of nasal cell gene expression. METHODS: The Wisconsin Infant Study Cohort included farm (n = 156) and nonfarm (n = 155) families with children followed to age 2 years. Parents reported prenatal farm and other environmental exposures. Illness frequency and severity were assessed using illness diaries and periodic surveys. Nasopharyngeal cell gene expression in a subset of 64 children at age 2 years was compared to farm exposure and respiratory illness history. RESULTS: Farm versus nonfarm children had nominally lower rates of respiratory illnesses (rate ratio 0.82 [95% CI, 0.69, 0.97]) with a stepwise reduction in illness rates in children exposed to 0, 1, or ≥2 animal species, but these trends were nonsignificant in a multivariable model. Farm exposures and preceding respiratory illnesses were positively related to nasal cell gene signatures for mononuclear cells and innate and antimicrobial responses. CONCLUSIONS: Maternal and infant exposure to farms and farm animals was associated with nonsignificant trends for reduced respiratory illnesses. Nasal cell gene expression in a subset of children suggests that farm exposures and respiratory illnesses in early life are associated with distinct patterns of mucosal immune expression.


Subject(s)
Environmental Exposure , Farms , Nasal Mucosa , Respiratory Tract Diseases , Humans , Female , Animals , Male , Infant , Environmental Exposure/adverse effects , Child, Preschool , Nasal Mucosa/immunology , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/epidemiology , Respiratory Tract Diseases/genetics , Animals, Domestic/immunology , Infant, Newborn , Wisconsin/epidemiology
8.
J Virol ; 97(11): e0132223, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37882519

ABSTRACT

IMPORTANCE: Chickens immunized with the infectious laryngotracheitis chicken embryo origin (CEO) vaccine (Medivac, PT Medion Farma Jaya) experience adverse reactions, hindering its safety and effective use in poultry flocks. To improve the effect of the vaccine, we sought to find a strategy to alleviate the respiratory reactions associated with the vaccine. Here, we confirmed that co-administering the CEO vaccine with chIL-2 by oral delivery led to significant alleviation of the vaccine reactions in chickens after immunization. Furthermore, we found that the co-administration of chIL-2 with the CEO vaccine reduced the clinical signs of the CEO vaccine while enhancing natural killer cells and cytotoxic T lymphocyte response to decrease viral loads in their tissues, particularly in the trachea and conjunctiva. Importantly, we demonstrated that the chIL-2 treatment can ameliorate the replication of the CEO vaccine without compromising its effectiveness. This study provides new insights into further applications of chIL-2 and a promising strategy for alleviating the adverse reaction of vaccines.


Subject(s)
Chickens , Herpesviridae Infections , Herpesvirus 1, Gallid , Interleukin-2 , Killer Cells, Natural , T-Lymphocytes, Cytotoxic , Viral Vaccines , Animals , Administration, Oral , Chickens/immunology , Chickens/virology , Conjunctiva/virology , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/veterinary , Herpesviridae Infections/virology , Herpesvirus 1, Gallid/immunology , Interleukin-2/administration & dosage , Interleukin-2/immunology , Killer Cells, Natural/immunology , Poultry Diseases/immunology , Poultry Diseases/prevention & control , Poultry Diseases/virology , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/prevention & control , Respiratory Tract Diseases/veterinary , Respiratory Tract Diseases/virology , T-Lymphocytes, Cytotoxic/immunology , Trachea/virology , Viral Load , Viral Vaccines/administration & dosage , Viral Vaccines/adverse effects , Viral Vaccines/biosynthesis , Viral Vaccines/immunology
9.
Int J Med Sci ; 20(6): 737-748, 2023.
Article in English | MEDLINE | ID: mdl-37213672

ABSTRACT

Purpose: The effectiveness of inactivated vaccines against acute respiratory syndrome coronavirus 2 (SARS­CoV­2), the causative agent of coronavirus disease 2019 (COVID-19), has become a global concern. Hence, the aim of this study was to evaluate vaccine safety and to assess immune responses in individuals with chronic respiratory disease (CRD) following a two-dose vaccination. Methods: The study cohort included 191 participants (112 adult CRD patients and 79 healthy controls [HCs]) at least 21 (range, 21-159) days after a second vaccination. Frequencies of memory B cells (MBCs) subsets and titers of SARS-CoV-2 neutralizing antibodies (NAbs) and anti-receptor binding domain (RBD) IgG antibodies (Abs) were analyzed. Results: As compared to the HCs, CRD patients had lower seropositivity rates and titers of both anti-RBD IgG Abs and NAbs, in addition to lower frequencies of RBD-specific MBCs (all, p < 0.05). At 3 months, CRD patients had lower seropositivity rates and titers of anti-RBD IgG Abs than the HCs (p < 0.05). For CoronaVac, the seropositivity rates of both Abs were lower in patients with old pulmonary tuberculosis than HCs. For BBIBP-CorV, the seropositivity rates of CoV-2 NAbs were lower in patients with chronic obstructive pulmonary disease than HCs (all, p < 0.05). Meanwhile, there was no significant difference in overall adverse events between the CRD patients and HCs. Univariate and multivariate analyses identified the time interval following a second vaccination as a risk factor for the production of anti-RBD IgG Abs and CoV-2 NAbs, while the CoronaVac had a positive effect on the titers of both Abs. Female was identified as a protective factor for CoV-2 NAb levels. Conclusion: Inactivated COVID-19 vaccines were safe and well tolerated by CRD patients but resulted in lower Ab responses and the frequencies of RBD-specific MBCs. Therefore, CRD patients should be prioritized for booster vaccinations.


Subject(s)
COVID-19 Vaccines , COVID-19 , Adult , Female , Humans , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , East Asian People , Immunity , Immunoglobulin G , SARS-CoV-2 , Vaccine Efficacy , Immunogenicity, Vaccine , Respiratory Tract Diseases/immunology , Chronic Disease
10.
Biomolecules ; 12(2)2022 01 28.
Article in English | MEDLINE | ID: mdl-35204727

ABSTRACT

The complement system (CS) is part of the human immune system, consisting of more than 30 proteins that play a vital role in the protection against various pathogens and diseases, including viral diseases. Activated via three pathways, the classical pathway (CP), the lectin pathway (LP), and the alternative pathway (AP), the complement system leads to the formation of a membrane attack complex (MAC) that disrupts the membrane of target cells, leading to cell lysis and death. Due to the increasing number of reports on its role in viral diseases, which may have implications for research on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), this review aims to highlight significant progress in understanding and defining the role of the complement system in four groups of diseases of viral etiology: (1) respiratory diseases; (2) acute liver failure (ALF); (3) disseminated intravascular coagulation (DIC); and (4) vector-borne diseases (VBDs). Some of these diseases already present a serious global health problem, while others are a matter of concern and require the collaboration of relevant national services and scientists with the World Health Organization (WHO) to avoid their spread.


Subject(s)
Complement System Proteins , Virus Diseases/etiology , Animals , Disseminated Intravascular Coagulation/immunology , Disseminated Intravascular Coagulation/virology , Humans , Liver Failure, Acute/immunology , Liver Failure, Acute/virology , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/virology , Vector Borne Diseases/immunology , Vector Borne Diseases/virology
11.
J Virol ; 96(5): e0172521, 2022 03 09.
Article in English | MEDLINE | ID: mdl-34985999

ABSTRACT

Influenza A virus (IAV) causes respiratory disease in swine and humans. Vaccines are used to prevent influenza illness in both populations but must be frequently updated due to rapidly evolving strains. Mismatch between the circulating strains and the strains contained in vaccines may cause loss of efficacy. Whole inactivated virus (WIV) vaccines with adjuvant, utilized by the swine industry, are effective against antigenically similar viruses; however, vaccine-associated enhanced respiratory disease (VAERD) may happen when the WIV is antigenically mismatched with the infecting virus. VAERD is a repeatable model in pigs, but had yet to be experimentally demonstrated in other mammalian species. We recapitulated VAERD in ferrets, a standard benchmark animal model for studying human influenza infection, in a direct comparison to VAERD in pigs. Both species were vaccinated with WIV with oil-in-water adjuvant containing a δ-1 H1N2 (1B.2.2) derived from the pre-2009 human seasonal lineage, then challenged with a 2009 pandemic H1N1 (H1N1pdm09, 1A.3.3.2) 5 weeks after vaccination. Nonvaccinated and challenged groups showed typical signs of influenza disease, but the mismatched vaccinated and challenged pigs and ferrets showed elevated clinical signs, despite similar viral loads. VAERD-affected pigs exhibited a 2-fold increase in lung lesions, while VAERD-affected ferrets showed a 4-fold increase. Similar to pigs, antibodies from VAERD-affected ferrets preferentially bound to the HA2 domain of the H1N1pdm09 challenge strain. These results indicate that VAERD is not limited to pigs, as demonstrated here in ferrets, and the need to consider VAERD when evaluating new vaccine platforms and strategies. IMPORTANCE We demonstrated the susceptibility of ferrets, a laboratory model species for human influenza A virus research, to vaccine-associated enhanced respiratory disease (VAERD) using an experimental model previously demonstrated in pigs. Ferrets developed clinical characteristics of VAERD very similar to that in pigs. The hemagglutinin (HA) stalk is a potential vaccine target to develop more efficacious, broadly reactive influenza vaccine platforms and strategies. However, non-neutralizing antibodies directed toward a conserved epitope on the HA stalk induced by an oil-in-water, adjuvanted, whole influenza virus vaccine were previously shown in VAERD-affected pigs and were also identified here in VAERD-affected ferrets. The induction of VAERD in ferrets highlights the potential risk of mismatched influenza vaccines for humans and the need to consider VAERD when designing and evaluating vaccine strategies.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Orthomyxoviridae Infections , Respiratory Tract Diseases , Animals , Antibodies, Viral , Disease Models, Animal , Ferrets , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/adverse effects , Influenza Vaccines/immunology , Influenza Vaccines/standards , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Respiratory Tract Diseases/immunology , Swine , Vaccines, Inactivated/immunology
12.
Viruses ; 13(12)2021 12 20.
Article in English | MEDLINE | ID: mdl-34960822

ABSTRACT

A microarray-based assay to detect IgG and IgM antibodies against betacoronaviruses (SARS-CoV-2, SARS, MERS, OC43, and HKU1), other respiratory viruses and type I interferons (IFN-Is) was developed. This multiplex assay was applied to track antibody cross-reactivity due to previous contact with similar viruses and to identify antibodies against IFN-Is as the markers for severe COVID-19. In total, 278 serum samples from convalescent plasma donors, COVID-19 patients in the intensive care unit (ICU) and patients who recovered from mild/moderate COVID-19, vaccine recipients, prepandemic and pandemic patients with autoimmune endocrine disorders, and a heterogeneous prepandemic cohort including healthy individuals and chronically ill patients were analyzed. The anti-SARS-CoV-2 microarray results agreed well with the ELISA results. Regarding ICU patients, autoantibodies against IFN-Is were detected in 10.5% of samples, and 10.5% of samples were found to simultaneously contain IgM antibodies against more than two different viruses. Cross-reactivity between IgG against the SARS-CoV-2 nucleocapsid and IgG against the OC43 and HKU1 spike proteins was observed, resulting in positive signals for the SARS-CoV-2 nucleocapsid in prepandemic samples from patients with autoimmune endocrine disorders. The presence of IgG against the SARS-CoV-2 nucleocapsid in the absence of IgG against the SARS-CoV-2 spike RBD should be interpreted with caution.


Subject(s)
Antibodies, Viral/immunology , Interferon Type I/immunology , SARS-CoV-2/immunology , Viruses/immunology , Antibodies, Viral/blood , Antigens, Viral/immunology , Autoantibodies/blood , Autoantibodies/immunology , COVID-19/immunology , COVID-19 Serological Testing , Cross Reactions , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Protein Array Analysis , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/virology , Viruses/classification
13.
Front Immunol ; 12: 733324, 2021.
Article in English | MEDLINE | ID: mdl-34630416

ABSTRACT

The lung is a vital mucosal organ that is constantly exposed to the external environment, and as such, its defenses are continuously under threat. The pulmonary immune system has evolved to sense and respond to these danger signals while remaining silent to innocuous aeroantigens. The origin of the defense system is the respiratory epithelium, which responds rapidly to insults by the production of an array of mediators that initiate protection by directly killing microbes, activating tissue-resident immune cells and recruiting leukocytes from the blood. At the steady-state, the lung comprises a large collection of leukocytes, amongst which are specialized cells of lymphoid origin known as innate lymphoid cells (ILCs). ILCs are divided into three major helper-like subsets, ILC1, ILC2 and ILC3, which are considered the innate counterparts of type 1, 2 and 17 T helper cells, respectively, in addition to natural killer cells and lymphoid tissue inducer cells. Although ILCs represent a small fraction of the pulmonary immune system, they play an important role in early responses to pathogens and facilitate the acquisition of adaptive immunity. However, it is now also emerging that these cells are active participants in the development of chronic lung diseases. In this mini-review, we provide an update on our current understanding of the role of ILCs and their regulation in the lung. We summarise how these cells and their mediators initiate, sustain and potentially control pulmonary inflammation, and their contribution to the respiratory diseases chronic obstructive pulmonary disease (COPD) and asthma.


Subject(s)
Lung/immunology , Lymphocytes/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Respiratory Tract Diseases/immunology , T-Lymphocyte Subsets/immunology , Adaptive Immunity , Animals , Chronic Disease , Humans , Immunity, Innate
14.
mSphere ; 6(3)2021 05 19.
Article in English | MEDLINE | ID: mdl-34011681

ABSTRACT

Staphylococcus aureus is both a commensal and a pathogenic bacterium for humans. Its ability to induce severe infections is based on a wide range of virulence factors. S. aureus community-acquired pneumonia (SA-CAP) is rare and severe, and the contribution of certain virulence factors in this disease has been recognized over the past 2 decades. First, the factors involved in metabolism adaptation are crucial for S. aureus survival in the lower respiratory tract, and toxins and enzymes are required for it to cross the pulmonary epithelial barrier. S. aureus subsequently faces host defense mechanisms, including the epithelial barrier, but most importantly the immune system. Here, again, S. aureus uses myriad virulence factors to successfully escape from the host's defenses and takes advantage of them. The impact of S. aureus virulence, combined with the collateral damage caused by an overwhelming immune response, leads to severe tissue damage and adverse clinical outcomes. In this review, we summarize step by step all of the S. aureus factors implicated in CAP and described to date, and we provide an outlook for future research.


Subject(s)
Pneumonia, Bacterial/immunology , Respiratory Tract Diseases/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Animals , Community-Acquired Infections/immunology , Community-Acquired Infections/microbiology , Humans , Mice , Pneumonia, Bacterial/pathology , Respiratory Tract Diseases/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Virulence , Virulence Factors
16.
Viruses ; 13(4)2021 04 16.
Article in English | MEDLINE | ID: mdl-33923693

ABSTRACT

Respiratory syncytial virus (RSV) infects most infants by two years of age. It can cause severe disease leading to an increased risk of developing asthma later in life. Previously, our group has shown that RSV infection in mice and infants promotes IL-1ß production. Here, we characterized the role of NLRP3-Inflammasome activation during RSV infection in adult mice and neonates. We observed that the inhibition of NLRP3 activation using the small molecule inhibitor, MCC950, or in genetically modified NLRP3 knockout (Nlrp3-/-) mice during in vivo RSV infection led to decreased lung immunopathology along with a reduced expression of the mucus-associated genes and reduced production of innate cytokines (IL-1ß, IL-33 and CCL2) linked to severe RSV disease while leading to significant increases in IFN-ß. NLRP3-inflammasome inhibition or deletion diminished Th2 cytokines and inflammatory cell infiltration into the lungs. Furthermore, NLRP3 inhibition or deletion during early-life RSV infection led to reducing viral-exacerbated allergic response in a mouse model of RSV-induced allergy exacerbation. Here, we demonstrated the critical role of NLRP3-inflammasome activation in RSV immunopathology and the related long-term airway alteration. Moreover, these findings suggest the NLRP3-inflammasome as a potential therapeutic target to attenuate severe RSV disease and limit childhood asthma development.


Subject(s)
Inflammasomes/antagonists & inhibitors , Lung/immunology , Lung/virology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Tract Diseases/prevention & control , Animals , Animals, Newborn , Cytokines/immunology , Female , Furans/administration & dosage , Indenes/administration & dosage , Inflammasomes/genetics , Inflammasomes/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Respiratory Syncytial Virus, Human/immunology , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/virology , Sulfonamides/administration & dosage
17.
Viruses ; 13(3)2021 02 24.
Article in English | MEDLINE | ID: mdl-33668216

ABSTRACT

Equine herpesvirus-1 is the cause of respiratory disease, abortion, and equine herpesvirus myeloencephalopathy (EHM) in horses worldwide. EHM affects as many as 14% of infected horses and a cell-associated viremia is thought to be central for EHM pathogenesis. While EHM is infrequent in younger horses, up to 70% of aged horses develop EHM. The aging immune system likely contributes to EHM pathogenesis; however, little is known about the host factors associated with clinical EHM. Here, we used the "old mare model" to induce EHM following EHV-1 infection. Peripheral blood mononuclear cells (PBMCs) of horses prior to infection and during viremia were collected and RNA sequencing with differential gene expression was used to compare the transcriptome of horses that did (EHM group) and did not (non-EHM group) develop clinical EHM. Interestingly, horses exhibiting EHM did not show respiratory disease, while non-EHM horses showed significant respiratory disease starting on day 2 post infection. Multiple immune pathways differed in EHM horses in response to EHV-1. These included an upregulation of IL-6 gene expression, a dysregulation of T-cell activation through AP-1 and responses skewed towards a T-helper 2 phenotype. Further, a dysregulation of coagulation and an upregulation of elements in the progesterone response were observed in EHM horses.


Subject(s)
Herpesviridae Infections/virology , Herpesvirus 1, Equid/immunology , Horse Diseases/immunology , Horse Diseases/virology , Leukocytes, Mononuclear/virology , Transcriptome/genetics , Animals , Female , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Profiling/methods , Herpesviridae Infections/immunology , Horses , Interleukin-6/genetics , Interleukin-6/immunology , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Respiratory Tract Diseases/genetics , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/virology , T-Lymphocytes/immunology , T-Lymphocytes/virology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology , Transcriptome/immunology , Up-Regulation/genetics , Up-Regulation/immunology
18.
Immunobiology ; 226(2): 152071, 2021 03.
Article in English | MEDLINE | ID: mdl-33588306

ABSTRACT

Dendritic cell-associated C-type lectin-1 (Dectin-1), also known as ß-glucan receptor is an emerging pattern recognition receptor (PRR) which belongs to the family of C-type lectin receptor (CLR). This CLR identifies ligands independently of Ca2+ and is majorly involved in coupling of innate with adaptive immunity. Formerly, Dectin-1 was best known for its role in anti-fungal defense only. However, recent explorations suggested its wider role in defense against variety of infectious diseases caused by pathogens including bacteria, parasites and viruses. In fact, Dectin-1 signaling axis has been suggested to be targeted as an effective therapeutic strategy for cancers. Dectin-1 has also been elucidated ascetically in the heart, respiratory, intestinal, neurological and developmental disorders. Being a defensive PRR, Dectin-1 results in optimal immune responses in collaboration with other PRRs, but the overall evaluation reinforces the hypothesis of disease development on dis-regulation of Dectin-1 activity. This underscores the impact of Dectin-1 polymorphisms in modulating protein expression and generation of non-optimal immune responses through defective collaborations, further underlining their therapeutic potential. To add on, Dectin-1 influence autoimmunity and severe inflammation accredited to recognition of self T cells and apoptotic cells through unknown ligands. Few reports have also testified its redundant role in infections, which makes it a complicated molecule to be fully resolved. Thus, Dectin-1 is a hub that runs a complex collaborative network, whose interactive wire connections to different PRRs are still pending to be revealed. Alternatively, so far focus of almost all the researchers was the two major cell surface isoforms of Dectin-1, despite the fact that its soluble functional intracellular isoform (Dectin-1E) has already been dissected but is indefinable. Therefore, this review intensely recommends the need of future research to resolve the un-resolved and treasure the comprehensive role of Dectin-1 in different clinical outcomes, before determining its therapeutic prospective.


Subject(s)
Lectins, C-Type/immunology , Receptors, Pattern Recognition/immunology , Animals , Autoimmune Diseases/immunology , Autophagy , Heart Diseases/immunology , Humans , Infections/immunology , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Neoplasms/immunology , Nervous System Diseases/immunology , Receptors, Pattern Recognition/chemistry , Receptors, Pattern Recognition/genetics , Respiratory Tract Diseases/immunology
19.
Life Sci ; 267: 118973, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33400932

ABSTRACT

Eosinophils are bi-lobed, multi-functional innate immune cells with diverse cell surface receptors that regulate local immune and inflammatory responses. Several inflammatory and infectious diseases are triggered with their build up in the blood and tissues. The mobilization of eosinophils into the lungs is regulated by a cascade of processes guided by Th2 cytokine generating T-cells. Recruitment of eosinophils essentially leads to a characteristic immune response followed by airway hyperresponsiveness and remodeling, which are hallmarks of chronic respiratory diseases. By analysing the dynamic interactions of eosinophils with their extracellular environment, which also involve signaling molecules and tissues, various therapies have been invented and developed to target respiratory diseases. Having entered clinical testing, several eosinophil targeting therapeutic agents have shown much promise and have further bridged the gap between theory and practice. Moreover, researchers now have a clearer understanding of the roles and mechanisms of eosinophils. These factors have successfully assisted molecular biologists to block specific pathways in the growth, migration and activation of eosinophils. The primary purpose of this review is to provide an overview of the eosinophil biology with a special emphasis on potential pharmacotherapeutic targets. The review also summarizes promising eosinophil-targeting agents, along with their mechanisms and rationale for use, including those in developmental pipeline, in clinical trials, or approved for other respiratory disorders.


Subject(s)
Eosinophils/immunology , Respiration Disorders/immunology , Respiratory Tract Diseases/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Eosinophils/drug effects , Eosinophils/metabolism , Humans , Lung/metabolism , Lung/pathology , Respiration Disorders/metabolism , Respiration Disorders/physiopathology , Respiratory Tract Diseases/metabolism , Respiratory Tract Diseases/physiopathology , Th2 Cells/immunology , Th2 Cells/metabolism
20.
Pediatr Res ; 90(5): 1023-1030, 2021 11.
Article in English | MEDLINE | ID: mdl-33504970

ABSTRACT

BACKGROUND: In infants admitted to an ICU with respiratory failure, there is an association between the ratio of CD8+ to CD4+ T cells within the upper respiratory tract and disease severity. Whether this ratio is associated with respiratory disease severity within children presenting to a pediatric emergency department is not known. METHODS: We studied a convenience sample of 63 children presenting to a pediatric emergency department with respiratory symptoms. T cell subsets in the nasal mucosa were analyzed by flow cytometry. We compared CD4+ and CD8+ T cells subsets in these samples and analyzed the proportion of these subsets that expressed markers associated with tissue residency. RESULTS: We were able to identify major subsets of CD8 and CD4 T cells within the nasal mucosa using flocked swabs. We found no difference in the ratio CD8+ to CD4+ T cells in children with upper or lower respiratory illness. A positive association between tissue-resident memory T cell frequency and patient age was identified. CONCLUSIONS: In our patient populations, the CD8+:CD4+ ratio was not associated with disease severity. The majority of T cells collected on nasal swabs are antigen experienced, and there is an association between the frequency of tissue-resident T cells and age. IMPACT: Immune cell populations from the nasal mucosa can be captured using flocked nasal swabs and analyzed by flow cytometry. Nasal CD8+:CD4+ ratio does not predict respiratory illness severity in children presenting to the emergency department. The frequency of CD8+ and CD4+ resident memory T cells within the nasal mucosa increases with age.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Nasal Mucosa/immunology , Respiratory Tract Diseases/immunology , T-Lymphocyte Subsets , Adolescent , Child , Child, Preschool , Cross-Sectional Studies , Female , Humans , Infant , Male
SELECTION OF CITATIONS
SEARCH DETAIL