Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.264
Filter
1.
Virologie (Montrouge) ; 28(4): 1-20, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39248668

ABSTRACT

While antiretroviral therapy (ART) has revolutionized the management of human immunodeficiency virus (HIV) and has enabled people living with HIV (PLWH) to achieve near-normal life expectancies, an HIV cure remains elusive due to the presence of HIV reservoirs. Furthermore, compared with individuals in the general population, PLWH support a higher burden of multimorbidity, including pulmonary diseases of both an infectious and non-infection nature, which may be a consequence of the formation of HIV reservoirs. Their gut, lymph nodes, brain, testes and lungs constitute important anatomic sites for the reservoirs. While CD4+ T cells, and particularly memory CD4+ T cells, are the best characterized cellular HIV reservoirs, tissue resident macrophages (TRM) and alveolar macrophages (AM) also harbor HIV infection. AM are the most abundant cells in bronchoalveolar (BAL) fluid in healthy conditions, and act as sentinels in the alveolar space by patrolling and clearing debris, microbes and surfactant recycling. Long-lived tissue-resident AM of embryonic origin have the capacity of self-renewal without replenishment from peripheral monocytes. As in other tissues, close cell-cell contacts in lungs also provide a milieu conducive for cell-to-cell spread of HIV infection and establishment of reservoirs. As lungs are in constant exposure to antigens from the external environment, this situation contributes to pro-inflammatory phenotype rendering pulmonary immune cells exhausted and senescent-an environment facilitating HIV persistence. Factors such as tobacco and e-cigarette smoking, lung microbiome dysbiosis and respiratory coinfections further drive antigenic stimulation and HIV replication. HIV replication, in turn, contributes to ongoing inflammation and clonal expansion. Herein, the potential role of AM in HIV persistence is discussed. Furthermore, their contribution towards pulmonary inflammation and immune dysregulation, which may in turn render PLWH susceptible to chronic lung disease, despite ART, is explored. Finally, strategies to eliminate HIV-infected AM are discussed.


Subject(s)
HIV Infections , Lung Diseases , Macrophages, Alveolar , Humans , HIV Infections/immunology , HIV Infections/virology , HIV Infections/complications , Macrophages, Alveolar/virology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/physiology , Lung Diseases/virology , Lung Diseases/immunology , Lung/virology , Lung/immunology , HIV-1/physiology , Disease Reservoirs/virology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology
2.
Virologie (Montrouge) ; 28(4): 255-276, 2024 Aug 01.
Article in French | MEDLINE | ID: mdl-39248670

ABSTRACT

While antiretroviral therapy (ART) has revolutionized the management of human immunodeficiency virus (HIV) and has enabled people living with HIV (PLWH) to achieve near-normal life expectancies, an HIV cure remains elusive due to the presence of HIV reservoirs. Furthermore, compared with individuals in the general population, PLWH support a higher burden of multimorbidity, including pulmonary diseases of both an infectious and non-infection nature, which may be a consequence of the formation of HIV reservoirs. Their gut, lymph nodes, brain, testes and lungs constitute important anatomic sites for the reservoirs. While CD4+ T-cells, and particularly memory CD4+ T-cells, are the best characterized cellular HIV reservoirs, tissue resident macrophages (TRM) and alveolar macrophages (AM) also harbor HIV infection. AM are the most abundant cells in bronchoalveolar (BAL) fluid in healthy conditions, and act as sentinels in the alveolar space by patrolling and clearing debris, microbes and surfactant recycling. Long-lived tissue-resident AM of embryonic origin have the capacity of self-renewal without replenishment from peripheral monocytes. As in other tissues, close cell-cell contacts in lungs also provide a milieu conducive for cell-to-cell spread of HIV infection and establishment of reservoirs. As lungs are in constant exposure to antigens from the external environment, this situation contributes to pro-inflammatory phenotype rendering pulmonary immune cells exhausted and senescent-an environment facilitating HIV persistence. Factors such as tobacco and e-cigarette smoking, lung microbiome dysbiosis and respiratory co-infections further drive antigenic stimulation and HIV replication. HIV replication, in turn, contributes to ongoing inflammation and clonal expansion. Herein, the potential role of AM in HIV persistence is discussed. Furthermore, their contribution towards pulmonary inflammation and immune dysregulation, which may in turn render PLWH susceptible to chronic lung disease, despite ART, is explored. Finally, strategies to eliminate HIV-infected AM are discussed.


Subject(s)
HIV Infections , Lung Diseases , Macrophages, Alveolar , Humans , HIV Infections/immunology , HIV Infections/virology , HIV Infections/complications , HIV Infections/drug therapy , Macrophages, Alveolar/virology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/physiology , Lung Diseases/virology , Lung Diseases/immunology , HIV-1/physiology , Lung/virology , Lung/immunology , Disease Reservoirs/virology
3.
BMC Pulm Med ; 24(1): 392, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138424

ABSTRACT

BACKGROUND: The immunologic features of nontuberculous mycobacterial pulmonary disease (NTM-PD) are largely unclear. This study investigated the immunologic features of NTM-PD using digital spatial profiling techniques. METHODS: Lung tissues obtained from six patients with NTM-PD between January 1, 2006, and December 31, 2020, at Seoul National University Hospital were subjected to RNA sequencing. Cores from the peribronchial areas were stained with CD3, CD68, and DNASyto13, and gene expression at the whole-transcriptome level was quantified using PCR amplification and Illumina sequencing. Lung tissues from six patients with bronchiectasis collected during the same period were used as controls. The RNA sequencing results were validated using immunohistochemistry (IHC) in another cohort (30 patients with NTM-PD and 15 patients with bronchiectasis). RESULTS: NTM-PD exhibited distinct gene expression patterns in T cells and macrophages. Gene set enrichment analysis revealed that pathways related to antigen presentation and processing were upregulated in NTM-PD, particularly in macrophages. Macrophages were more prevalent and the expression of genes associated with the M1 phenotype (CD40 and CD80) was significantly elevated. Although macrophages were activated in the NTM-PD group T cell activity was unaltered. Notably, expression of the costimulatory molecule CD28 was decreased in NTM-PD. IHC analysis showed that T cells expressing Foxp3 or TIM-3, which facilitate the regulatory functions of T cells, were increased. CONCLUSIONS: NTM-PD exhibits distinct immunologic signatures characterized by the activation of macrophages without T cell activation.


Subject(s)
Mycobacterium Infections, Nontuberculous , Humans , Male , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium Infections, Nontuberculous/genetics , Female , Middle Aged , Aged , Transcriptome , Macrophages/immunology , Macrophages/metabolism , Lung/microbiology , Lung/immunology , Lung/pathology , Nontuberculous Mycobacteria/genetics , Nontuberculous Mycobacteria/immunology , Lung Diseases/genetics , Lung Diseases/microbiology , Lung Diseases/immunology , T-Lymphocytes/immunology , Gene Expression Profiling , Adult , Bronchiectasis/immunology , Bronchiectasis/genetics , Bronchiectasis/microbiology
4.
Front Immunol ; 15: 1427739, 2024.
Article in English | MEDLINE | ID: mdl-39026681

ABSTRACT

Lactylation is a process where lactate, a cellular metabolism byproduct, is added to proteins, altering their functions. In the realm of macrophage activation, lactylation impacts inflammatory response and immune regulation. Understanding the effects of lactylation on macrophage activation is vital in lung diseases, as abnormal activation and function are pivotal in conditions like pneumonia, pulmonary fibrosis, COPD, and lung cancer. This review explores the concept of lactylation, its regulation of macrophage activation, and recent research progress in lung diseases. It offers new insights into lung disease pathogenesis and potential therapeutic targets.


Subject(s)
Lung Diseases , Macrophage Activation , Humans , Macrophage Activation/immunology , Animals , Lung Diseases/immunology , Lung Diseases/metabolism , Lactic Acid/metabolism , Macrophages/immunology , Macrophages/metabolism
5.
Front Immunol ; 15: 1405376, 2024.
Article in English | MEDLINE | ID: mdl-39015565

ABSTRACT

Pseudomonas aeruginosa is a highly adaptable opportunistic pathogen capable of exploiting barriers and immune defects to cause chronic lung infections in conditions such as cystic fibrosis. In these contexts, host immune responses are ineffective at clearing persistent bacterial infection, instead driving a cycle of inflammatory lung damage. This review outlines key components of the host immune response to chronic P. aeruginosa infection within the lung, beginning with initial pathogen recognition, followed by a robust yet maladaptive innate immune response, and an ineffective adaptive immune response that propagates lung damage while permitting bacterial persistence. Untangling the interplay between host immunity and chronic P. aeruginosa infection will allow for the development and refinement of strategies to modulate immune-associated lung damage and potentiate the immune system to combat chronic infection more effectively.


Subject(s)
Host-Pathogen Interactions , Immunity, Innate , Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Pseudomonas aeruginosa/immunology , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Chronic Disease , Animals , Host-Pathogen Interactions/immunology , Adaptive Immunity , Lung Diseases/immunology , Lung Diseases/microbiology , Cystic Fibrosis/immunology , Cystic Fibrosis/microbiology , Cystic Fibrosis/complications , Lung/immunology , Lung/microbiology
6.
Physiol Rev ; 104(4): 1643-1677, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39052018

ABSTRACT

Representing severe morbidity and mortality globally, respiratory infections associated with chronic respiratory diseases, including complicated pneumonia, asthma, interstitial lung disease, and chronic obstructive pulmonary disease, are a major public health concern. Lung health and the prevention of pulmonary disease rely on the mechanisms of airway surface fluid secretion, mucociliary clearance, and adequate immune response to eradicate inhaled pathogens and particulate matter from the environment. The antimicrobial proteins and peptides contribute to maintaining an antimicrobial milieu in human lungs to eliminate pathogens and prevent them from causing pulmonary diseases. The predominant antimicrobial molecules of the lung environment include human α- and ß-defensins and cathelicidins, among numerous other host defense molecules with antimicrobial and antibiofilm activity such as PLUNC (palate, lung, and nasal epithelium clone) family proteins, elafin, collectins, lactoferrin, lysozymes, mucins, secretory leukocyte proteinase inhibitor, surfactant proteins SP-A and SP-D, and RNases. It has been demonstrated that changes in antimicrobial molecule expression levels are associated with regulating inflammation, potentiating exacerbations, pathological changes, and modifications in chronic lung disease severity. Antimicrobial molecules also display roles in both anticancer and tumorigenic effects. Lung antimicrobial proteins and peptides are promising alternative therapeutics for treating and preventing multidrug-resistant bacterial infections and anticancer therapies.


Subject(s)
Antimicrobial Peptides , Lung , Humans , Animals , Lung/immunology , Lung/metabolism , Lung Diseases/drug therapy , Lung Diseases/immunology , Lung Diseases/metabolism , Antimicrobial Cationic Peptides/metabolism
7.
Curr Allergy Asthma Rep ; 24(8): 415-423, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38904934

ABSTRACT

PURPOSE OF REVIEW: The human commensal microbiota is now widely accepted as a key regulator of human health and disease. The composition of the mucosal associated microbiota has been shown to play a critical role in the lung health. The role of the mucosal microbiota in the development and severity of allergy, asthma, and occupational lung disease is only beginning to take shape. However, advances in our understanding of these links have tremendous potential to led to new clinical interventions to reduce allergy, asthma, and occupational lung disease morbidity. RECENT FINDINGS: We review recent work describing the relationship and role of the commensal microbiota in the development of allergy, asthma, and occupational lung disease. Our review primarily focuses on occupational exposures and the effects of the microbiome, both in composition and function. Data generated from these studies may lead to the development of interventions targeted at establishing and maintaining a healthy microbiota. We also highlight the role of environmental exposures and the effects on the commensal microbial community and their potential association with occupational lung disease. This review explores the current research describing the role of the human microbiome in the regulation of pulmonary health and disease, with a specific focus on the role of the mucosal microbiota in the development of allergy, asthma, and occupational lung disease.


Subject(s)
Asthma , Hypersensitivity , Microbiota , Occupational Diseases , Humans , Microbiota/immunology , Asthma/immunology , Asthma/microbiology , Hypersensitivity/immunology , Hypersensitivity/microbiology , Occupational Diseases/microbiology , Occupational Diseases/immunology , Occupational Exposure/adverse effects , Lung Diseases/microbiology , Lung Diseases/immunology
8.
Clin Lab ; 70(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38868881

ABSTRACT

BACKGROUND: IgG4-related disease (IgG4-RD) is an immune-mediated systemic inflammatory fibrotic disease, which is a relatively rare and novel disease that can involve multiple organs or tissues, with variable clinical manifestations, and for which pulmonary involvement has been reported relatively infrequently. METHODS: Here we report a case of pulmonary infection that was initially suspected and received anti-inflammatory treatment, but the symptoms did not improve. CT examination indicated progression of the pulmonary lesion, and the nature of the lesion could not be determined by tracheoscopy and bronchoalveolar lavage. The diagnosis of IgG4 related lung disease (IgG4-RLD) was confirmed by percutaneous lung biopsy. A joint literature analysis was conducted to improve clinicians' understanding of this disease. RESULTS: The patient's history, symptoms, signs and relevant examination results were analyzed. The final diagnosis was IgG4-RLD. CONCLUSIONS: When the clinical symptoms and imaging manifestations of the patients are consistent with IgG4-RLD, pathological examination can be appropriately performed to clarify the nature of the lesions. More consideration should be given to the possibility of disease diagnosis to avoid misdiagnosis and underdiagnosis, and proper treatment should be given at an early stage.


Subject(s)
Immunoglobulin G4-Related Disease , Immunoglobulin G , Lung Diseases , Tomography, X-Ray Computed , Humans , Immunoglobulin G4-Related Disease/diagnosis , Immunoglobulin G4-Related Disease/immunology , Lung Diseases/diagnosis , Lung Diseases/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Lung/diagnostic imaging , Lung/pathology , Lung/immunology , Middle Aged , Biopsy
9.
Int J Mol Sci ; 25(10)2024 May 12.
Article in English | MEDLINE | ID: mdl-38791316

ABSTRACT

Pulmonary manifestations of vasculitis are associated with significant morbidity and mortality in affected individuals. They result from a complex interplay between immune dysregulation, which leads to vascular inflammation and tissue damage. This review explored the underlying pathogenesis of pulmonary involvement in vasculitis, encompassing various forms such as granulomatosis with polyangiitis (GPA), microscopic polyangiitis (MPA), eosinophilic granulomatosis with polyangiitis (EGPA), and anti-GBM disease. Mechanisms involving ANCA and anti-GBM autoantibodies, neutrophil activation, and neutrophil extracellular trap (NETs) formation are discussed, along with the role of the complement system in inducing pulmonary injury. Furthermore, the impact of genetic predisposition and environmental factors on disease susceptibility and severity was considered, and the current treatment options were presented. Understanding the mechanisms involved in the pathogenesis of pulmonary vasculitis is crucial for developing targeted therapies and improving clinical outcomes in affected individuals.


Subject(s)
Anti-Glomerular Basement Membrane Disease , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis , Humans , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/immunology , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/pathology , Anti-Glomerular Basement Membrane Disease/immunology , Anti-Glomerular Basement Membrane Disease/etiology , Anti-Glomerular Basement Membrane Disease/pathology , Extracellular Traps/metabolism , Extracellular Traps/immunology , Antibodies, Antineutrophil Cytoplasmic/immunology , Lung Diseases/etiology , Lung Diseases/immunology , Lung Diseases/pathology , Lung/pathology , Lung/immunology , Autoantibodies/immunology , Animals , Microscopic Polyangiitis/immunology , Microscopic Polyangiitis/complications , Microscopic Polyangiitis/pathology
10.
Front Immunol ; 15: 1383503, 2024.
Article in English | MEDLINE | ID: mdl-38756780

ABSTRACT

With the development of global social economy and the deepening of the aging population, diseases related to aging have received increasing attention. The pathogenesis of many respiratory diseases remains unclear, and lung aging is an independent risk factor for respiratory diseases. The aging mechanism of the lung may be involved in the occurrence and development of respiratory diseases. Aging-induced immune, oxidative stress, inflammation, and telomere changes can directly induce and promote the occurrence and development of lung aging. Meanwhile, the occurrence of lung aging also further aggravates the immune stress and inflammatory response of respiratory diseases; the two mutually affect each other and promote the development of respiratory diseases. Explaining the mechanism and treatment direction of these respiratory diseases from the perspective of lung aging will be a new idea and research field. This review summarizes the changes in pulmonary microenvironment, metabolic mechanisms, and the progression of respiratory diseases associated with aging.


Subject(s)
Aging , Cellular Microenvironment , Lung , Oxidative Stress , Humans , Aging/immunology , Lung/immunology , Animals , Lung Diseases/immunology , Lung Diseases/etiology , Inflammation/immunology
12.
Sheng Li Xue Bao ; 76(2): 346-352, 2024 Apr 25.
Article in Chinese | MEDLINE | ID: mdl-38658383

ABSTRACT

Programmed death-ligand 1 (PD-L1) is important in maintaining central and peripheral immune tolerance in normal tissues, mediating tumor immune escape and keeping the balance between anti- and pro-inflammatory responses. Inflammation plays an important role in inflammatory lung diseases. This article reviews the research progress and potential clinical value of PD-L1 in inflammatory lung diseases, including acute lung injury, chronic obstructive pulmonary disease, asthma and idiopathic pulmonary fibrosis.


Subject(s)
Asthma , B7-H1 Antigen , Pulmonary Disease, Chronic Obstructive , Humans , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Asthma/immunology , Acute Lung Injury/immunology , Inflammation/immunology , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/metabolism , Lung Diseases/immunology , Lung Diseases/metabolism , Animals
13.
Arch. bronconeumol. (Ed. impr.) ; 60(3): 161-170, Mar. 2024. tab, ilus
Article in English | IBECS | ID: ibc-231100

ABSTRACT

Respiratory syncytial virus (RSV) is a major cause of respiratory tract infections in adults, particularly older adults and those with underlying medical conditions. Vaccination has emerged as a potential key strategy to prevent RSV-related morbidity and mortality. This Neumoexperts Prevention (NEP) Group scientific paper aims to provide an evidence-based positioning and RSV vaccination recommendations for adult patients. We review the current literature on RSV burden and vaccine development and availability, emphasising the importance of vaccination in the adult population. According to our interpretation of the data, RSV vaccines should be part of the adult immunisation programme, and an age-based strategy should be preferred over targeting high-risk groups. The effectiveness and efficiency of this practice will depend on the duration of protection and the need for annual or more spaced doses. Our recommendations should help healthcare professionals formulate guidelines and implement effective vaccination programmes for adult patients at risk of RSV infection now that specific vaccines are available.(AU)


Subject(s)
Humans , Male , Female , Middle Aged , Aged , Respiratory Syncytial Viruses/immunology , Vaccination , Disease Prevention , Lung Diseases/prevention & control , Lung Diseases/immunology , Immunization Programs
14.
Mucosal Immunol ; 17(3): 359-370, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38492745

ABSTRACT

Suppurative lung disease and wheezing are common respiratory diseases of childhood, however, due to poor understanding of underlying pathobiology, there are limited treatment options and disease recurrence is common. We aimed to profile the pulmonary and systemic immune response in children with wheeze and chronic suppurative lung disease for identification of endotypes that can inform improved clinical management. We used clinical microbiology data, highly multiplexed flow cytometry and immunoassays to compare pulmonary [bronchoalveolar lavage (BAL)] and systemic immunity in children with lung disease and controls. Unsupervised analytical approaches were applied to BAL immune data to explore biological endotypes. We identified two endotypes that were analogous in both frequency and immune signature across both respiratory diseases. The hyper-inflammatory endotype had a 12-fold increase in neutrophil infiltration and upregulation of 14 soluble signatures associated with type 2 inflammation and cell recruitment to tissue. The non-inflammatory endotype was not significantly different from controls. We showed these endotypes are measurable in a clinical setting and can be defined by measuring only three immune factors in BAL. We identified hyper-inflammatory and non-inflammatory endotypes common across pediatric wheeze and chronic suppurative lung disease that, if validated in future studies, have the potential to inform clinical management.


Subject(s)
Bronchoalveolar Lavage Fluid , Respiratory Sounds , Humans , Respiratory Sounds/immunology , Male , Female , Child , Child, Preschool , Bronchoalveolar Lavage Fluid/immunology , Lung/immunology , Lung/pathology , Lung Diseases/immunology , Lung Diseases/etiology , Inflammation/immunology , Infant , Cytokines/metabolism , Adolescent , Biomarkers
15.
Allergy ; 79(5): 1089-1122, 2024 05.
Article in English | MEDLINE | ID: mdl-38108546

ABSTRACT

The accumulation of senescent cells drives inflammaging and increases morbidity of chronic inflammatory lung diseases. Immune responses are built upon dynamic changes in cell metabolism that supply energy and substrates for cell proliferation, differentiation, and activation. Metabolic changes imposed by environmental stress and inflammation on immune cells and tissue microenvironment are thus chiefly involved in the pathophysiology of allergic and other immune-driven diseases. Altered cell metabolism is also a hallmark of cell senescence, a condition characterized by loss of proliferative activity in cells that remain metabolically active. Accelerated senescence can be triggered by acute or chronic stress and inflammatory responses. In contrast, replicative senescence occurs as part of the physiological aging process and has protective roles in cancer surveillance and wound healing. Importantly, cell senescence can also change or hamper response to diverse therapeutic treatments. Understanding the metabolic pathways of senescence in immune and structural cells is therefore critical to detect, prevent, or revert detrimental aspects of senescence-related immunopathology, by developing specific diagnostics and targeted therapies. In this paper, we review the main changes and metabolic alterations occurring in senescent immune cells (macrophages, B cells, T cells). Subsequently, we present the metabolic footprints described in translational studies in patients with chronic asthma and chronic obstructive pulmonary disease (COPD), and review the ongoing preclinical studies and clinical trials of therapeutic approaches aiming at targeting metabolic pathways to antagonize pathological senescence. Because this is a recently emerging field in allergy and clinical immunology, a better understanding of the metabolic profile of the complex landscape of cell senescence is needed. The progress achieved so far is already providing opportunities for new therapies, as well as for strategies aimed at disease prevention and supporting healthy aging.


Subject(s)
Cellular Senescence , Metabolic Networks and Pathways , Humans , Cellular Senescence/drug effects , Animals , Chronic Disease , Inflammation/metabolism , Inflammation/immunology , Lung Diseases/etiology , Lung Diseases/drug therapy , Lung Diseases/metabolism , Lung Diseases/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/immunology , Aging/immunology , Aging/metabolism
16.
Int J Mol Sci ; 24(23)2023 Nov 21.
Article in English | MEDLINE | ID: mdl-38068879

ABSTRACT

Inflammation and inflammasomes have been proposed as important regulators of the host-microorganism interaction, playing a key role in morbidity and mortality due to the coronavirus disease 2019 (COVID-19) in subjects with chronic conditions and compromised immune system. The inflammasome consists of a multiprotein complex that finely regulates the activation of caspase-1 and the production and secretion of potent pro-inflammatory cytokines such as IL-1ß and IL-18. The pyrin containing NOD (nucleotide-binding oligomerization domain) like receptor (NLRP) is a family of intracellular receptors, sensing patterns associated to pathogens or danger signals and NLRP3 inflammasome is the most deeply analyzed for its involvement in the innate and adaptive immune system as well as its contribution to several autoinflammatory and autoimmune diseases. It is highly expressed in leukocytes and up-regulated in sentinel cells upon inflammatory stimuli. NLRP3 expression has also been reported in B and T lymphocytes, in epithelial cells of oral and genital mucosa, in specific parenchymal cells as cardiomyocytes, and keratinocytes, and chondrocytes. It is well known that a dysregulated activation of the inflammasome is involved in the pathogenesis of different disorders that share the common red line of inflammation in their pathogenetic fingerprint. Here, we review the potential roles of the NLRP3 inflammasome in cardiovascular events, liver damage, pulmonary diseases, and in that wide range of systemic inflammatory syndromes named as a cytokine storm.


Subject(s)
Cytokine Release Syndrome , Heart Diseases , Inflammasomes , Liver Diseases , Lung Diseases , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Carrier Proteins/metabolism , Cytokine Release Syndrome/immunology , Inflammasomes/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Lung Diseases/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Heart Diseases/immunology , Liver Diseases/immunology
17.
J Biol Chem ; 299(12): 105365, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37865317

ABSTRACT

Glycan structure is often modulated in disease or predisease states, suggesting that such changes might serve as biomarkers. Here, we generated a monoclonal antibody (mAb) against the core fucose of the N-glycan in human IgG. Notably, this mAb can be used in Western blotting and ELISA. ELISA using this mAb revealed a low level of the core fucose of the N-glycan in IgG, suggesting that the level of acore fucosylated (noncore fucosylated) IgG was increased in the sera of the patients with lung cancer, chronic obstructive pulmonary disease, and interstitial pneumonia compared to healthy subjects. In a coculture analysis using human lung adenocarcinoma A549 cells and antibody-secreting B cells, the downregulation of the FUT8 (α1,6 fucosyltransferase) gene and a low level of core fucose of the N-glycan in IgG in antibody-secreting B cells were observed after coculture. A dramatic alteration in gene expression profiles for cytokines, chemokines, and their receptors were also observed after coculturing, and we found that the identified C-C motif chemokine 2 was partially involved in the downregulation of the FUT8 gene and the low level of core fucose of the N-glycan in IgG in antibody-secreting B cells. We also developed a latex turbidimetric immunoassay using this mAb. These results suggest that communication with C-C motif chemokine 2 between lung cells and antibody-secreting B cells downregulate the level of core fucose of the N-glycan in IgG, i.e., the increased level of acore fucosylated (noncore fucosylated) IgG, which would be a novel biomarker for the diagnosis of patients with pulmonary diseases.


Subject(s)
Antibodies, Monoclonal , Fucose , Immunoglobulin G , Lung Diseases , Polysaccharides , Humans , A549 Cells , Antibodies, Monoclonal/metabolism , Antibody Specificity , B-Lymphocytes/immunology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokines/genetics , Chemokines/metabolism , Fucose/blood , Fucose/metabolism , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Gene Expression Profiling , Gene Expression Regulation/immunology , Gene Knockout Techniques , Immunoassay/standards , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lung Diseases/diagnosis , Lung Diseases/immunology , Polysaccharides/metabolism , Animals , Mice , CHO Cells , HEK293 Cells , Cricetulus
18.
Toxicol Lett ; 383: 141-151, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37394155

ABSTRACT

Long-term inhalation of silica nanoparticles (SiNPs) can induce pulmonary fibrosis (PF), nevertheless, the potential mechanisms remain elusive. Herein, we constructed a three-dimensional (3D) co-culture model by using Matrigel to investigate the interaction among different cells and potential regulatory mechanisms after SiNPs exposure. Methodologically, we dynamically observed the changes in cell morphology and migration after exposure to SiNPs by co-culturing mouse monocytic macrophages (RAW264.7), human non-small cell lung cancer cells (A549), and medical research council cell strain-5 (MRC-5) in Matrigel for 24 h. Subsequently, we detected the expression of nuclear factor kappa B (NF-κB), inflammatory factor and epithelial-mesenchymal transition (EMT) markers. The results showed that SiNPs produced toxic effects on cells. In the 3D co-culture state, the cell's movement velocity and displacement increased, and the cell migration ability was enhanced. Meanwhile, the expression of inflammatory factor tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) were upregulated, the epithelial marker E-cadherin (E-cad) was downregulated, the mesenchymal marker N-cadherin (N-cad) and myofibroblast marker alpha-smooth muscle actin (α-SMA) expression were upregulated, while NF-κB expression was also upregulated after SiNPs exposure. We further found that cells were more prone to transdifferentiate into myofibroblasts in the 3D co-culture state. Conversely, utilizing the NF-κB-specific inhibitor BAY 11-7082 effectively downregulated the expression of TNF-α, IL-6, interleukin-1ß (IL-1ß), N-cad, α-SMA, collagen-I (COL I), and fibronectin (FN), the expression of E-cad was upregulated. These findings suggest that NF-κB is involved in regulating SiNPs-induced inflammatory, EMT, and fibrosis in the 3D co-culture state.


Subject(s)
Epithelial-Mesenchymal Transition , Fibrosis , Lung Diseases , Nanoparticles , Silicon Dioxide , Animals , Humans , Mice , Carcinoma, Non-Small-Cell Lung , Coculture Techniques , Epithelial-Mesenchymal Transition/immunology , Fibrosis/etiology , Fibrosis/immunology , Interleukin-6 , Lung Neoplasms , Nanoparticles/toxicity , NF-kappa B/metabolism , Silicon Dioxide/toxicity , Tumor Necrosis Factor-alpha/metabolism , Lung Diseases/etiology , Lung Diseases/immunology
19.
Microbiol Spectr ; 10(6): e0311022, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36255321

ABSTRACT

Nontuberculous mycobacteria (NTM) cause pulmonary disease in individuals without obvious immunodeficiency. This study was initiated to gain insight into the immunological factors that predispose persons to NTM pulmonary disease (NTMPD). Blood was obtained from 15 pairs of NTMPD patients and their healthy household contacts. Peripheral blood mononuclear cells (PBMCs) were stimulated with the Mycobacterium avium complex (MAC). A total of 34 cytokines and chemokines were evaluated in plasma and PBMC culture supernatants using multiplex immunoassays, and gene expression in the PBMCs was determined using real-time PCR. PBMCs from NTMPD patients produced significantly less interleukin-1ß (IL-1ß), IL-18, IL-1α, and IL-10 than PBMCs from their healthy household contacts in response to MAC. Although plasma RANTES levels were high in NTMPD patients, they had no effect on IL-1ß production by macrophages infected with MAC. Toll-like receptor 2 (TLR2) and TWIK2 (a two-pore domain K+ channel) were impaired in response to MAC in PBMCs of NTMPD patients. A TLR2 inhibitor decreased all four cytokines, whereas a two-pore domain K+ channel inhibitor decreased the production of IL-1ß, IL-18, and IL-1α, but not IL-10, by MAC-stimulated PBMCs and monocytes. The ratio of monocytes was reduced in whole blood of NTMPD patients compared with that of healthy household contacts. A reduced monocyte ratio might contribute to the attenuated production of IL-1 family cytokines by PBMCs of NTMPD patients in response to MAC stimulations. Collectively, our findings suggest that the attenuated IL-1 response may increase susceptibility to NTM pulmonary infection through multiple factors, including impaired expression of the TLR2 and TWIK2 and reduced monocyte ratio. IMPORTANCE Upon MAC stimulation, the production of IL-1 family cytokines and IL-10 by PBMCs of NTMPD patients was attenuated compared with that of healthy household contacts. Upon MAC stimulation, the expression of TLR2 and TWIK2 (one of the two-pore domain K+ channels) was attenuated in PBMCs of NTMPD patients compared with that of healthy household contacts. The production of IL-1 family cytokines by MAC-stimulated PBMCs and MAC-infected monocytes of healthy donors was reduced by a TLR2 inhibitor and two-pore domain K+ channel inhibitor. The ratio of monocytes was reduced in whole blood of NTMPD patients compared with that of healthy household contacts. Collectively, our data suggest that defects in the expression of TLR2 and TWIK2 in human PBMCs or monocytes and reduced monocyte ratio are involved in the reduced production of IL-1 family cytokines, and it may increase susceptibility to NTM pulmonary infection.


Subject(s)
Cytokines , Lung Diseases , Mycobacterium Infections, Nontuberculous , Pneumonia, Bacterial , Humans , Interleukin-18/immunology , Leukocytes, Mononuclear , Lung Diseases/immunology , Monocytes/immunology , Mycobacterium avium Complex , Mycobacterium Infections, Nontuberculous/immunology , Toll-Like Receptor 2/immunology , Pneumonia, Bacterial/immunology , Cytokines/immunology
20.
J Immunol ; 208(5): 1180-1188, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35149529

ABSTRACT

Pulmonary infections elicit a combination of tissue-resident and circulating T cell responses. Understanding the contribution of these anatomically distinct cellular pools in protective immune responses is critical for vaccine development. Francisella tularensis is a highly virulent bacterium capable of causing lethal systemic disease following pulmonary infection for which there is no currently licensed vaccine. Although T cells are required for survival of F. tularensis infection, the relative contribution of tissue-resident and circulating T cells is not completely understood, hampering design of effective, long-lasting vaccines directed against this bacterium. We have previously shown that resident T cells were not sufficient to protect against F. tularensis, suggesting circulating cells may serve a critical role in host defense. To elucidate the role of circulating T cells, we used a model of vaccination and challenge of parabiotic mice. Intranasally infected naive mice conjoined to immune animals had increased numbers of circulating memory T cells and similar splenic bacterial burdens as vaccinated-vaccinated pairs. However, bacterial loads in the lungs of naive parabionts were significantly greater than those observed in vaccinated-vaccinated pairs, but despite early control of F. tularensis replication, all naive-vaccinated pairs succumbed to infection. Together, these data define the specific roles of circulating and resident T cells in defense against infection that is initiated in the pulmonary compartment but ultimately causes disseminated disease. These data also provide evidence for employing vaccination strategies that elicit both pools of T cells for immunity against F. tularensis and may be a common theme for other disseminating bacterial infections.


Subject(s)
Bacterial Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Francisella tularensis/immunology , Memory T Cells/immunology , Animals , Antibodies, Bacterial/blood , Bacterial Load/immunology , Female , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Lung Diseases/immunology , Lung Diseases/microbiology , Lung Diseases/pathology , Mice , Mice, Inbred C57BL , Tularemia/immunology , Tularemia/pathology , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL