Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.545
Filter
1.
Respir Res ; 25(1): 193, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702733

ABSTRACT

BACKGROUND: Influenza A virus (IAV) infection is a significant risk factor for respiratory diseases, but the host defense mechanisms against IAV remain to be defined. Immune regulators such as surfactant protein A (SP-A) and Toll-interacting protein (Tollip) have been shown to be involved in IAV infection, but whether SP-A and Tollip cooperate in more effective host defense against IAV infection has not been investigated. METHODS: Wild-type (WT), Tollip knockout (KO), SP-A KO, and Tollip/SP-A double KO (dKO) mice were infected with IAV for four days. Lung macrophages were isolated for bulk RNA sequencing. Precision-cut lung slices (PCLS) from WT and dKO mice were pre-treated with SP-A and then infected with IAV for 48 h. RESULTS: Viral load was significantly increased in bronchoalveolar lavage (BAL) fluid of dKO mice compared to all other strains of mice. dKO mice had significantly less recruitment of neutrophils into the lung compared to Tollip KO mice. SP-A treatment of PCLS enhanced expression of TNF and reduced viral load in dKO mouse lung tissue. Pathway analysis of bulk RNA sequencing data suggests that macrophages from IAV-infected dKO mice reduced expression of genes involved in neutrophil recruitment, IL-17 signaling, and Toll-like receptor signaling. CONCLUSIONS: Our data suggests that both Tollip and SP-A are essential for the lung to exert more effective innate defense against IAV infection.


Subject(s)
Influenza A virus , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections , Pulmonary Surfactant-Associated Protein A , Animals , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein A/genetics , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/metabolism , Influenza A virus/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Lung/immunology , Lung/metabolism , Lung/virology
2.
Front Immunol ; 15: 1382638, 2024.
Article in English | MEDLINE | ID: mdl-38715601

ABSTRACT

Recovery from respiratory pneumococcal infections generates lung-localized protection against heterotypic bacteria, mediated by resident memory lymphocytes. Optimal protection in mice requires re-exposure to pneumococcus within days of initial infection. Serial surface marker phenotyping of B cell populations in a model of pneumococcal heterotypic immunity revealed that bacterial re-exposure stimulates the immediate accumulation of dynamic and heterogeneous populations of B cells in the lung, and is essential for the establishment of lung resident memory B (BRM) cells. The B cells in the early wave were activated, proliferating locally, and associated with both CD4+ T cells and CXCL13. Antagonist- and antibody-mediated interventions were implemented during this early timeframe to demonstrate that lymphocyte recirculation, CD4+ cells, and CD40 ligand (CD40L) signaling were all needed for lung BRM cell establishment, whereas CXCL13 signaling was not. While most prominent as aggregates in the loose connective tissue of bronchovascular bundles, morphometry and live lung imaging analyses showed that lung BRM cells were equally numerous as single cells dispersed throughout the alveolar septae. We propose that CD40L signaling from antigen-stimulated CD4+ T cells in the infected lung is critical to establishment of local BRM cells, which subsequently protect the airways and parenchyma against future potential infections.


Subject(s)
CD4-Positive T-Lymphocytes , CD40 Ligand , Lung , Memory B Cells , Streptococcus pneumoniae , Animals , CD40 Ligand/metabolism , CD40 Ligand/immunology , Mice , Streptococcus pneumoniae/immunology , Lung/immunology , Lung/pathology , Lung/microbiology , CD4-Positive T-Lymphocytes/immunology , Memory B Cells/immunology , Memory B Cells/metabolism , Pneumococcal Infections/immunology , Mice, Inbred C57BL , Immunologic Memory , Chemokine CXCL13/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Disease Models, Animal , Signal Transduction , Lymphocyte Activation/immunology
3.
Front Immunol ; 15: 1382655, 2024.
Article in English | MEDLINE | ID: mdl-38803494

ABSTRACT

Introduction: Global microplastic (MP) pollution is now well recognized, with humans and animals consuming and inhaling MPs on a daily basis, with a growing body of concern surrounding the potential impacts on human health. Methods: Using a mouse model of mild COVID-19, we describe herein the effects of azide-free 1 µm polystyrene MP beads, co-delivered into lungs with a SARS-CoV-2 omicron BA.5 inoculum. The effect of MPs on the host response to SARS-CoV-2 infection was analysed using histopathology and RNA-Seq at 2 and 6 days post-infection (dpi). Results: Although infection reduced clearance of MPs from the lung, virus titres and viral RNA levels were not significantly affected by MPs, and overt MP-associated clinical or histopathological changes were not observed. However, RNA-Seq of infected lungs revealed that MP exposure suppressed innate immune responses at 2 dpi and increased pro-inflammatory signatures at 6 dpi. The cytokine profile at 6 dpi showed a significant correlation with the 'cytokine release syndrome' signature observed in some COVID-19 patients. Discussion: The findings are consistent with the recent finding that MPs can inhibit phagocytosis of apoptotic cells via binding of Tim4. They also add to a growing body of literature suggesting that MPs can dysregulate inflammatory processes in specific disease settings.


Subject(s)
COVID-19 , Disease Models, Animal , Immunity, Innate , Lung , Microplastics , SARS-CoV-2 , Animals , COVID-19/immunology , COVID-19/virology , Immunity, Innate/drug effects , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Mice , Lung/immunology , Lung/virology , Lung/pathology , Cytokines/metabolism , Humans , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Female , Cytokine Release Syndrome/immunology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Betacoronavirus/immunology , Pandemics
4.
Clin Transl Sci ; 17(6): e13850, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38807464

ABSTRACT

Cold-inducible RNA-binding protein (CIRP) is a damage-associated molecular pattern that plays a critical role in triggering inflammatory responses. It remains unknown whether CIRP is strongly associated with bacterial load, inflammatory response, and mortality in sepsis model. Pneumonia was induced in specific pathogen-free 8-9-week old male rats by injecting bacteria via puncture of the tracheal cartilage. The expressions of CIRP and proinflammatory cytokines [tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and IL-1ß] in lung tissues, alveolar macrophages (AMs), plasma, and bronchoalveolar lavage fluid (BALF) were determined by reverse transcription-polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assay. The numbers of bacteria recovered from the lungs were correlated with the bacterial loads injected and mortality. The expressions of CIRP increased sharply as the bacterial loads increased in the lung tissues and AMs. The amounts of TNF-α, IL-6 and IL-1ß proteins synthesized were dependent on the bacterial load in the lung tissues. Releases of CIRP, TNF-α, IL-6, and IL-1ß increased with the bacterial load in the blood plasma. The proteins confirmed similar patterns in the BALF. CIRP was strongly associated with the releases of TNF-α, IL-6, and IL-1ß in the lung tissues, blood plasma, and BALF, and showed a close correlation with mortality. CIRP demonstrated a strong association with bacterial load, which is new evidence, and close correlations with proinflammatory cytokines and mortality of pneumonia in rats, suggesting that it might be an interesting pneumonic biomarker for monitoring host response and predicting mortality, and a promising target for immunotherapy.


Subject(s)
Bacterial Load , Cytokines , RNA-Binding Proteins , Animals , Male , RNA-Binding Proteins/metabolism , Cytokines/metabolism , Cytokines/blood , Rats , Lung/microbiology , Lung/immunology , Lung/pathology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Pneumonia/microbiology , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/mortality , Rats, Sprague-Dawley , Interleukin-1beta/metabolism , Interleukin-1beta/blood , Disease Models, Animal , Inflammation Mediators/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/blood , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/mortality
5.
Front Immunol ; 15: 1384697, 2024.
Article in English | MEDLINE | ID: mdl-38807596

ABSTRACT

Background: Asthma is a common obstructive airway disease with an inflammatory etiology. The main unmet need in the management of asthma is inadequate adherence to pharmacotherapy, leading to a poorly-controlled disease state, necessitating the development of novel therapies. Bronchom is a calcio-herbal formulation, which is purported to treat chronic asthma. The objective of the current study was to examine the in-vivo efficacy of Bronchom in mouse model of allergic asthma. Methods: Ultra high performance liquid chromatography was utilized to analyze the phytocompounds in Bronchom. Further, the in-vivo efficacy of Bronchom was evaluated in House dust mite (HDM)-induced allergic asthma in mice. Mice were challenged with aerosolized methacholine to assess airway hyperresponsiveness. Subsequently, inflammatory cell influx was evaluated in bronchoalveolar lavage fluid (BALF) followed by lung histology, wherein airway remodeling features were studied. Simultaneously, the levels of Th2 cytokines and chemokines in the BALF was also evaluated. Additionally, the mRNA expression of pro-inflammatory and Th2 cytokines was also assessed in the lung along with the oxidative stress markers. Results: Phytocompounds present in Bronchom included, gallic acid, protocatechuic acid, methyl gallate, rosmarinic acid, glycyrrhizin, eugenol, 6-gingerol and piperine. Bronchom effectively suppressed HDM-induced airway hyperresponsiveness along with the influx of leukocytes in the BALF. Additionally, Bronchom reduced the infiltration of inflammatory cells in the lung and it also ameliorated goblet cell metaplasia, sub-epithelial fibrosis and increase in α-smooth muscle actin. Bronchom decreased Th2 cytokines (IL-4 and IL-5) and chemokines (Eotaxin and IP-10) in the BALF. Likewise, it could also suppress the mRNA expression of pro-inflammatory cytokines (TNF-α, IFN-γ, IL-6 and IL-33), and IL-13. Moreover, Bronchom restored the HDM-induced diminution of endogenous anti-oxidants (GSH and SOD) and the increase in pro-oxidants (GSSG and MDA). Furthermore, Bronchom could also decrease the nitrosative stress by lowering the observed increase in nitrite levels. Conclusion: Taken together, the results of the present study data convincingly demonstrate that Bronchom exhibits pharmacological effects in an animal model of allergic asthma. Bronchom mitigated airway hyperresponsiveness, inflammation and airway remodeling evoked by a clinically relevant allergen and accordingly it possesses therapeutic potential for the treatment of asthma.


Subject(s)
Asthma , Chemokines , Cytokines , Disease Models, Animal , Goblet Cells , Metaplasia , Pyroglyphidae , Th2 Cells , Animals , Asthma/drug therapy , Asthma/immunology , Mice , Cytokines/metabolism , Goblet Cells/pathology , Goblet Cells/immunology , Goblet Cells/drug effects , Pyroglyphidae/immunology , Th2 Cells/immunology , Chemokines/metabolism , Fibrosis , Mice, Inbred BALB C , Airway Remodeling/drug effects , Female , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Lung/pathology , Lung/immunology , Lung/drug effects
6.
Front Immunol ; 15: 1374541, 2024.
Article in English | MEDLINE | ID: mdl-38807598

ABSTRACT

Objective: The coronavirus disease 2019 (COVID-19) spread rapidly and claimed millions of lives worldwide. Acute respiratory distress syndrome (ARDS) is the major cause of COVID-19-associated deaths. Due to the limitations of current drugs, developing effective therapeutic options that can be used rapidly and safely in clinics for treating severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infections is necessary. This study aims to investigate the effects of two food-extracted immunomodulatory agents, ajoene-enriched garlic extract (AGE) and cruciferous vegetables-extracted sulforaphane (SFN), on anti-inflammatory and immune responses in a SARS-CoV-2 acute lung injury mouse model. Methods: In this study, we established a mouse model to mimic the SARS-CoV-2 infection acute lung injury model via intratracheal injection of polyinosinic:polycytidylic acid (poly[I:C]) and SARS-CoV-2 recombinant spike protein (SP). After the different agents treatment, lung sections, bronchoalveolar lavage fluid (BALF) and fresh faeces were harvested. Then, H&E staining was used to examine symptoms of interstitial pneumonia. Flow cytometry was used to examine the change of immune cell populations. Multiplex cytokines assay was used to examine the inflammatory cytokines.16S rDNA high-throughput sequencing was used to examine the change of gut microbiome. Results: Our results showed that AGE and SFN significantly suppressed the symptoms of interstitial pneumonia, effectively inhibited the production of inflammatory cytokines, decreased the percentage of inflammatory cell populations, and elevated T cell populations in the mouse model. Furthermore, we also observed that the gut microbiome of genus Paramuribaculum were enriched in the AGE-treated group. Conclusion: Here, for the first time, we observed that these two novel, safe, and relatively inexpensive immunomodulatory agents exhibited the same effects on anti-inflammatory and immune responses as neutralizing monoclonal antibodies (mAbs) against interleukin 6 receptor (IL-6R), which have been suggested for treating COVID-19 patients. Our results revealed the therapeutic ability of these two immunomodulatory agents in a mouse model of SARS-CoV-2 acute lung injury by promoting anti-inflammatory and immune responses. These results suggest that AGE and SFN are promising candidates for the COVID-19 treatment.


Subject(s)
Acute Lung Injury , Angiotensin-Converting Enzyme 2 , Anti-Inflammatory Agents , COVID-19 Drug Treatment , COVID-19 , Disease Models, Animal , Immunomodulating Agents , SARS-CoV-2 , Animals , Mice , Acute Lung Injury/immunology , Acute Lung Injury/drug therapy , Acute Lung Injury/etiology , COVID-19/immunology , SARS-CoV-2/immunology , Immunomodulating Agents/pharmacology , Immunomodulating Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Isothiocyanates/pharmacology , Isothiocyanates/therapeutic use , Sulfoxides , Humans , Cytokines/metabolism , Spike Glycoprotein, Coronavirus/immunology , Lung/immunology , Lung/pathology , Lung/virology , Lung/drug effects , Male , Poly I-C , Plant Extracts/pharmacology , Plant Extracts/therapeutic use
7.
Immunohorizons ; 8(5): 384-396, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38809232

ABSTRACT

The mammalian Siglec receptor sialoadhesin (Siglec1, CD169) confers innate immunity against the encapsulated pathogen group B Streptococcus (GBS). Newborn lung macrophages have lower expression levels of sialoadhesin at birth compared with the postnatal period, increasing their susceptibility to GBS infection. In this study, we investigate the mechanisms regulating sialoadhesin expression in the newborn mouse lung. In both neonatal and adult mice, GBS lung infection reduced Siglec1 expression, potentially delaying acquisition of immunity in neonates. Suppression of Siglec1 expression required interactions between sialic acid on the GBS capsule and the inhibitory host receptor Siglec-E. The Siglec1 gene contains multiple STAT binding motifs, which could regulate expression of sialoadhesin downstream of innate immune signals. Although GBS infection reduced STAT1 expression in the lungs of wild-type newborn mice, we observed increased numbers of STAT1+ cells in Siglece-/- lungs. To test if innate immune activation could increase sialoadhesin at birth, we first demonstrated that treatment of neonatal lung macrophages ex vivo with inflammatory activators increased sialoadhesin expression. However, overcoming the low sialoadhesin expression at birth using in vivo prenatal exposures or treatments with inflammatory stimuli were not successful. The suppression of sialoadhesin expression by GBS-Siglec-E engagement may therefore contribute to disease pathogenesis in newborns and represent a challenging but potentially appealing therapeutic opportunity to augment immunity at birth.


Subject(s)
Animals, Newborn , Mice, Knockout , N-Acetylneuraminic Acid , STAT1 Transcription Factor , Sialic Acid Binding Ig-like Lectin 1 , Streptococcal Infections , Streptococcus agalactiae , Animals , Mice , Streptococcus agalactiae/immunology , N-Acetylneuraminic Acid/metabolism , Sialic Acid Binding Ig-like Lectin 1/metabolism , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Immunity, Innate , Mice, Inbred C57BL , Lung/immunology , Lung/microbiology , Lung/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Female , Macrophages/immunology , Macrophages/metabolism , Lectins/metabolism , Lectins/genetics , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Antigens, CD/metabolism , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte
8.
Front Immunol ; 15: 1372957, 2024.
Article in English | MEDLINE | ID: mdl-38779688

ABSTRACT

Background: Schistosomiasis is a common cause of pulmonary hypertension (PH) worldwide. Type 2 inflammation contributes to the development of Schistosoma-induced PH. Specifically, interstitial macrophages (IMs) derived from monocytes play a pivotal role by producing thrombospondin-1 (TSP-1), which in turn activates TGF-ß, thereby driving the pathology of PH. Resident and recruited IM subpopulations have recently been identified. We hypothesized that in Schistosoma-PH, one IM subpopulation expresses monocyte recruitment factors, whereas recruited monocytes become a separate IM subpopulation that expresses TSP-1. Methods: Mice were intraperitoneally sensitized and then intravenously challenged with S. mansoni eggs. Flow cytometry on lungs and blood was performed on wildtype and reporter mice to identify IM subpopulations and protein expression. Single-cell RNA sequencing (scRNAseq) was performed on flow-sorted IMs from unexposed and at day 1, 3 and 7 following Schistosoma exposure to complement flow cytometry based IM characterization and identify gene expression. Results: Flow cytometry and scRNAseq both identified 3 IM subpopulations, characterized by CCR2, MHCII, and FOLR2 expression. Following Schistosoma exposure, the CCR2+ IM subpopulation expanded, suggestive of circulating monocyte recruitment. Schistosoma exposure caused increased monocyte-recruitment ligand CCL2 expression in the resident FOLR2+ IM subpopulation. In contrast, the vascular pathology-driving protein TSP-1 was greatest in the CCR2+ IM subpopulation. Conclusion: Schistosoma-induced PH involves crosstalk between IM subpopulations, with increased expression of monocyte recruitment ligands by resident FOLR2+ IMs, and the recruitment of CCR2+ IMs which express TSP-1 that activates TGF-ß and causes PH.


Subject(s)
Hypertension, Pulmonary , Macrophages , Animals , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/parasitology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/pathology , Mice , Macrophages/immunology , Macrophages/parasitology , Phenotype , Schistosoma mansoni/immunology , Mice, Inbred C57BL , Schistosomiasis/immunology , Schistosomiasis/complications , Schistosomiasis/parasitology , Disease Models, Animal , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Schistosomiasis mansoni/complications , Schistosomiasis mansoni/pathology , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Monocytes/immunology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Female , Schistosoma/immunology , Schistosoma/physiology , Lung/immunology , Lung/parasitology , Lung/pathology
9.
Mycoses ; 67(5): e13746, 2024 May.
Article in English | MEDLINE | ID: mdl-38767275

ABSTRACT

BACKGROUND: Post-tuberculosis lung abnormality (PTLA) is the most common risk factor for chronic pulmonary aspergillosis (CPA), and 14%-25% of the subjects with PTLA develop CPA. The pathogenesis and the host immune response in subjects with PTLA who develop CPA need to be better understood. METHODS: We prospectively compared the innate and adaptive immune responses mounted by patients of PTLA with or without CPA (controls). We studied the neutrophil oxidative burst (by dihydrorhodamine 123 test), classic (serum C3 and C4 levels) and alternative (mannose-binding lectin [MBL] protein levels) complement pathway, serum immunoglobulins (IgG, IgM and IgA), B and T lymphocytes and their subsets in subjects with PTLA with or without CPA. RESULTS: We included 111 subjects (58 CPA and 53 controls) in the current study. The mean ± SD age of the study population was 42.6 ± 15.7 years. The cases and controls were matched for age, gender distribution and body weight. Subjects with CPA had impaired neutrophil oxidative burst, lower memory T lymphocytes and impaired Th-1 immune response (lower Th-1 lymphocytes) than controls. We found no significant difference between the two groups in the serum complement levels, MBL levels, B-cell subsets and other T lymphocyte subsets. CONCLUSION: Subjects with CPA secondary to PTLA have impaired neutrophil oxidative burst and a lower Th-1 response than controls.


Subject(s)
Adaptive Immunity , Immunity, Innate , Pulmonary Aspergillosis , Tuberculosis, Pulmonary , Humans , Female , Male , Adult , Middle Aged , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/complications , Prospective Studies , Pulmonary Aspergillosis/immunology , Pulmonary Aspergillosis/complications , Neutrophils/immunology , Lung/immunology , Respiratory Burst , Young Adult
10.
Front Immunol ; 15: 1380628, 2024.
Article in English | MEDLINE | ID: mdl-38774866

ABSTRACT

Introduction: TAM receptor-mediated efferocytosis plays an important function in immune regulation and may contribute to antigen tolerance in the lungs, a site with continuous cellular turnover and generation of apoptotic cells. Some studies have identified failures in efferocytosis as a common driver of inflammation and tissue destruction in lung diseases. Our study is the first to characterize the in vivo function of the TAM receptors, Axl and MerTk, in the innate immune cell compartment, cytokine and chemokine production, as well as the alveolar macrophage (AM) phenotype in different settings in the airways and lung parenchyma. Methods: We employed MerTk and Axl defective mice to induce acute silicosis by a single exposure to crystalline silica particles (20 mg/50 µL). Although both mRNA levels of Axl and MerTk receptors were constitutively expressed by lung cells and isolated AMs, we found that MerTk was critical for maintaining lung homeostasis, whereas Axl played a role in the regulation of silica-induced inflammation. Our findings imply that MerTk and Axl differently modulated inflammatory tone via AM and neutrophil recruitment, phenotype and function by flow cytometry, and TGF-ß and CXCL1 protein levels, respectively. Finally, Axl expression was upregulated in both MerTk-/- and WT AMs, confirming its importance during inflammation. Conclusion: This study provides strong evidence that MerTk and Axl are specialized to orchestrate apoptotic cell clearance across different circumstances and may have important implications for the understanding of pulmonary inflammatory disorders as well as for the development of new approaches to therapy.


Subject(s)
Axl Receptor Tyrosine Kinase , Homeostasis , Lung , Macrophages, Alveolar , Mice, Knockout , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases , Silicosis , c-Mer Tyrosine Kinase , Animals , c-Mer Tyrosine Kinase/metabolism , c-Mer Tyrosine Kinase/genetics , Silicosis/metabolism , Silicosis/immunology , Silicosis/pathology , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Mice , Lung/immunology , Lung/metabolism , Lung/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice, Inbred C57BL , Cytokines/metabolism , Disease Models, Animal
11.
Front Immunol ; 15: 1365174, 2024.
Article in English | MEDLINE | ID: mdl-38774873

ABSTRACT

Introduction: Sepsis remains a major source of morbidity and mortality in neonates, and characterization of immune regulation in the neonatal septic response remains limited. HVEM is a checkpoint regulator which can both stimulate or inhibit immune responses and demonstrates altered expression after sepsis. We hypothesized that signaling via HVEM would be essential for the neonatal response to sepsis, and that therefore blockade of this pathway would improve survival to septic challenge. Methods: To explore this, neonatal mice were treated with cecal slurry (CS), CS with Anti-HVEM antibody (CS-Ab) or CS with isotype (CS-IT) and followed for 7-day survival. Mice from all treatment groups had thymus, lung, kidney and peritoneal fluid harvested, weighed, and stained for histologic evaluation, and changes in cardiac function were assessed with echocardiography. Results: Mortality was significantly higher for CS-Ab mice (72.2%) than for CS-IT mice (22.2%). CS resulted in dysregulated alveolar remodeling, but CS-Ab lungs demonstrated significantly less dysfunctional alveolar remodeling than CS alone (MCL 121.0 CS vs. 87.6 CS-Ab), as well as increased renal tubular vacuolization. No morphologic differences in alveolar septation or thymic karyorrhexis were found between CS-Ab and CS-IT. CS-Ab pups exhibited a marked decrease in heart rate (390.3 Sh vs. 342.1 CS-Ab), stroke volume (13.08 CS-IT vs. 8.83 CS-Ab) and ultimately cardiac output (4.90 Sh vs. 3.02 CS-Ab) as well as a significant increase in ejection fraction (73.74 Sh vs. 83.75 CS-Ab) and cardiac strain (40.74 Sh vs. 51.16 CS-Ab) as compared to CS-IT or Sham animals. Discussion: While receptor ligation of aspects of HVEM signaling, via antibody blockade, appears to mitigate aspects of lung injury and thymic involution, stimulatory signaling via HVEM still seems to be necessary for vascular and hemodynamic resilience and overall neonatal mouse survival in response to this experimental polymicrobial septic insult. This dissonance in the activity of anti-HVEM neutralizing antibody in neonatal animals speaks to the differences in how septic cardiac dysfunction should be considered and approached in the neonatal population.


Subject(s)
Animals, Newborn , Neonatal Sepsis , Signal Transduction , Animals , Mice , Neonatal Sepsis/immunology , Neonatal Sepsis/mortality , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Receptors, Tumor Necrosis Factor, Member 14/immunology , Disease Models, Animal , Female , Heart Diseases/etiology , Heart Diseases/immunology , Lung/immunology , Lung/pathology , Sepsis/immunology , Sepsis/metabolism
12.
Nat Commun ; 15(1): 4326, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773113

ABSTRACT

Resolving inflammation is thought to return the affected tissue back to homoeostasis but recent evidence supports a non-linear model of resolution involving a phase of prolonged immune activity. Here we show that within days following resolution of Streptococcus pneumoniae-triggered lung inflammation, there is an influx of antigen specific lymphocytes with a memory and tissue-resident phenotype as well as macrophages bearing alveolar or interstitial phenotype. The transcriptome of these macrophages shows enrichment of genes associated with prostaglandin biosynthesis and genes that drive T cell chemotaxis and differentiation. Therapeutic depletion of post-resolution macrophages, inhibition of prostaglandin E2 (PGE2) synthesis or treatment with an EP4 antagonist, MF498, reduce numbers of lung CD4+/CD44+/CD62L+ and CD4+/CD44+/CD62L-/CD27+ T cells as well as their expression of the α-integrin, CD103. The T cells fail to reappear and reactivate upon secondary challenge for up to six weeks following primary infection. Concomitantly, EP4 antagonism through MF498 causes accumulation of lung macrophages and marked tissue fibrosis. Our study thus shows that PGE2 signalling, predominantly via EP4, plays an important role during the second wave of immune activity following resolution of inflammation. This secondary immune activation drives local tissue-resident T cell development while limiting tissue injury.


Subject(s)
Dinoprostone , Disease Models, Animal , Lung , Macrophages , Mice, Inbred C57BL , Pneumonia, Pneumococcal , Receptors, Prostaglandin E, EP4 Subtype , Streptococcus pneumoniae , Animals , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/pathology , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/metabolism , Mice , Dinoprostone/metabolism , Streptococcus pneumoniae/immunology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Macrophages/immunology , Macrophages/metabolism , Lung/immunology , Lung/pathology , Lung/microbiology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Integrin alpha Chains/metabolism , Integrin alpha Chains/genetics , Female , Antigens, CD/metabolism , Antigens, CD/genetics , T-Lymphocytes/immunology
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(4): 296-302, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38710513

ABSTRACT

Objective To evaluate the effects of heme oxygenase-1 (HO-1) gene deletion on immune cell composition and inflammatory injury in lung tissues of mice with lipopolysaccharide (LPS)-induced acute lung injury (ALI). Methods C57BL/6 wild-type (WT) mice and HO-1 conditional-knockout (HO-1-/-) mice on the same background were randomly divided into four groups (n=5 in every group): WT control group, LPS-treated WT group, HO-1-/- control group and LPS-treated HO-1-/- group. LPS-treated WT and HO-1-/- groups were injected with LPS (15 mg/kg) through the tail vein to establish ALI model, while WT control group and HO-1-/- control group were injected with an equivalent volume of normal saline through the tail vein, respectively. Twelve hours later, the mice were sacrificed and lung tissues from each group were collected for analysis. Histopathological alterations of lung tissues were assessed by HE staining. The levels of mRNA expression of tumor necrosis factor α (TNF-α), interleukin 1ß (IL-1ß), and IL-6 were determined by PCR. The percentages of neutrophils (CD45+CD11b+Ly6G+Ly6C-), total monocytes (CD45+CD11b+Ly6Chi), pro-inflammatory monocyte subsets (CD45+CD11b+Ly6ChiCCR2hi) and total macrophages (CD45+CD11b+F4/80+), M1 macrophage (CD45+CD11b+F4/80+CD86+), M2 macrophage (CD45+CD11b+F4/80+CD206+), total T cells (CD45+CD3+), CD3+CD4+ T cells, CD3+CD8+ T cells and myeloid suppressor cells (MDSCs, CD45+CD11b+Gr1+) were detected by flow cytometry. Results Compared with the corresponding control groups, HE staining exhibited increased inflammation in the lung tissues of both LPS-treated WT and HO-1-/- model mice; mRNA expression levels of TNF-α, IL-1ß and IL-6 were up-regulated; the proportions of neutrophils, total monocytes, pro-inflammatory monocyte subsets, MDSCs and total macrophages increased significantly. The percentage of CD3+, CD3+CD4+ and CD3+CD8+ T cells decreased significantly. Under resting-state, compared with WT control mice, the proportion of neutrophils, monocytes and pro-inflammatory monocyte subset increased in lung tissues of HO-1-/- control mice, while the proportion of CD3+ and CD3+CD8+ T cells decreased. Compared with LPS-treated WT mice, the mRNA expression levels of TNF-α and IL-1ß were up-regulated in lung tissues of LPS-treated HO-1-/- mice; the proportion of total monocytes, pro-inflammatory monocyte subsets, M1 macrophages and M1/M2 ratio increased greatly; the percentage of CD3+CD8+ T cells decreased significantly. Conclusion The deletion of HO-1 affects the function of the lung immune system and aggravates the inflammatory injury after LPS stimulation in ALI mice.


Subject(s)
Acute Lung Injury , Heme Oxygenase-1 , Lipopolysaccharides , Lung , Mice, Inbred C57BL , Mice, Knockout , Animals , Male , Mice , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Inflammation/genetics , Inflammation/chemically induced , Inflammation/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Lung/pathology , Lung/immunology , Lung/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
Front Immunol ; 15: 1377374, 2024.
Article in English | MEDLINE | ID: mdl-38745662

ABSTRACT

Respiratory syncytial virus (RSV) is the primary cause of bronchiolitis-related hospitalizations among children under 5 years of age, with reinfection being common throughout life. Maternal vaccination has emerged as a promising strategy, delivering elevated antibody levels to newborns for immediate protection. However, limited research has explored the protective efficacy of maternal antibodies (matAbs) against secondary RSV infections in offspring. To address this gap, we employed a mouse model of maternal RSV vaccination and secondary infection of offspring to evaluate lung pathology following RSV reinfection in mice with varying levels of maternal antibody (matAb). Additionally, we aimed to investigate the potential causes of exacerbated lung inflammation in offspring with high matAb levels following secondary RSV exposure. Our findings revealed that offspring with elevated levels of maternal pre-F antibody demonstrated effective protection against lung pathology following the initial RSV infection. However, this protection was compromised upon reinfection, manifesting as heightened weight loss, exacerbated lung pathology, increased expression of RSV-A N genes, eosinophilia, enhanced IL-5, IL-13, MUC5AC, and eosinophils Major Basic Protein (MBP) production in lung tissue compared to offspring lacking matAbs. Importantly, these unexpected outcomes were not attributed to antibody-dependent enhancement (ADE) resulting from declining matAb levels over time. Notably, our findings showed a decline in secretory IgA (sIgA), mucosal IgA, and mucosal IgG levels in offspring with high matAb levels post-primary RSV challenge. We propose that this decline may be a critical factor contributing to the ineffective protection observed during secondary RSV exposure. Overall, these findings offer valuable insights into maternal vaccination against RSV, contributing to a comprehensive understanding and mitigation of potential risks associated with maternal RSV vaccination.


Subject(s)
Antibodies, Viral , Pneumonia , Respiratory Syncytial Virus Infections , Animals , Respiratory Syncytial Virus Infections/immunology , Mice , Female , Antibodies, Viral/blood , Antibodies, Viral/immunology , Pneumonia/immunology , Immunity, Maternally-Acquired , Lung/immunology , Lung/virology , Lung/pathology , Pregnancy , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/administration & dosage , Disease Models, Animal , Respiratory Syncytial Viruses/immunology , Mice, Inbred BALB C
15.
Front Immunol ; 15: 1372959, 2024.
Article in English | MEDLINE | ID: mdl-38690277

ABSTRACT

Introduction: Hypoxia is a common pathological driver contributing to various forms of pulmonary vascular diseases leading to pulmonary hypertension (PH). Pulmonary interstitial macrophages (IMs) play pivotal roles in immune and vascular dysfunction, leading to inflammation, abnormal remodeling, and fibrosis in PH. However, IMs' response to hypoxia and their role in PH progression remain largely unknown. We utilized a murine model of hypoxia-induced PH to investigate the repertoire and functional profiles of IMs in response to acute and prolonged hypoxia, aiming to elucidate their contributions to PH development. Methods: We conducted single-cell transcriptomic analyses to characterize the repertoire and functional profiles of murine pulmonary IMs following exposure to hypobaric hypoxia for varying durations (0, 1, 3, 7, and 21 days). Hallmark pathways from the mouse Molecular Signatures Database were utilized to characterize the molecular function of the IM subpopulation in response to hypoxia. Results: Our analysis revealed an early acute inflammatory phase during acute hypoxia exposure (Days 1-3), which was resolved by Day 7, followed by a pro-remodeling phase during prolonged hypoxia (Days 7-21). These phases were marked by distinct subpopulations of IMs: MHCIIhiCCR2+EAR2+ cells characterized the acute inflammatory phase, while TLF+VCAM1hi cells dominated the pro-remodeling phase. The acute inflammatory phase exhibited enrichment in interferon-gamma, IL-2, and IL-6 pathways, while the pro-remodeling phase showed dysregulated chemokine production, hemoglobin clearance, and tissue repair profiles, along with activation of distinct complement pathways. Discussion: Our findings demonstrate the existence of distinct populations of pulmonary interstitial macrophages corresponding to acute and prolonged hypoxia exposure, pivotal in regulating the inflammatory and remodeling phases of PH pathogenesis. This understanding offers potential avenues for targeted interventions, tailored to specific populations and distinct phases of the disease. Moreover, further identification of triggers for pro-remodeling IMs holds promise in unveiling novel therapeutic strategies for pulmonary hypertension.


Subject(s)
Gene Expression Profiling , Hypertension, Pulmonary , Hypoxia , Single-Cell Analysis , Transcriptome , Animals , Mice , Hypoxia/metabolism , Hypoxia/immunology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/genetics , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice, Inbred C57BL , Disease Models, Animal , Male , Lung/immunology , Lung/pathology , Lung/metabolism
16.
Front Immunol ; 15: 1342497, 2024.
Article in English | MEDLINE | ID: mdl-38694499

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a phenotypically heterogenous group of cells that potently suppress the immune response. A growing body of evidence supports the important role of MDSCs in a variety of lung diseases, such as asthma. However, the role of MDSCs in asthma exacerbation has so far not been investigated. Here, we studied the role of MDSCs in a murine model of influenza virus-induced asthma exacerbation. BALB/c mice were exposed to house dust mite (HDM) three times a week for a total of five weeks to induce a chronic asthmatic phenotype, which was exacerbated by additional exposure to the A/Hamburg/5/2009 hemagglutinin 1 neuraminidase 1 (H1N1) influenza virus. Induction of lung inflammatory features, production of T helper (Th) 1- and Th2- associated inflammatory cytokines in the lavage fluid and an increased airway hyper-responsiveness were observed, establishing the asthma exacerbation model. The number and activity of pulmonary M-MDSCs increased in exacerbated asthmatic mice compared to non-exacerbated asthmatic mice. Furthermore, depletion of MDSCs aggravated airway hyper-responsiveness in exacerbated asthmatic mice. These findings further denote the role of MDSCs in asthma and provide some of the first evidence supporting a potential important role of MDSCs in asthma exacerbation.


Subject(s)
Asthma , Cytokines , Disease Models, Animal , Influenza A Virus, H1N1 Subtype , Mice, Inbred BALB C , Myeloid-Derived Suppressor Cells , Orthomyxoviridae Infections , Animals , Asthma/immunology , Myeloid-Derived Suppressor Cells/immunology , Mice , Orthomyxoviridae Infections/immunology , Cytokines/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Female , Pyroglyphidae/immunology , Disease Progression , Lung/immunology , Lung/pathology , Lung/virology , Th2 Cells/immunology
17.
Front Immunol ; 15: 1360412, 2024.
Article in English | MEDLINE | ID: mdl-38745652

ABSTRACT

A robust immune response is required for resistance to pulmonary tuberculosis (TB), the primary disease caused by Mycobacterium tuberculosis (Mtb). However, pharmaceutical inhibition of T cell immune checkpoint molecules can result in the rapid development of active disease in latently infected individuals, indicating the importance of T cell immune regulation. In this study, we investigated the potential role of CD200R during Mtb infection, a key immune checkpoint for myeloid cells. Expression of CD200R was consistently downregulated on CD14+ monocytes in the blood of subjects with active TB compared to healthy controls, suggesting potential modulation of this important anti-inflammatory pathway. In homogenized TB-diseased lung tissue, CD200R expression was highly variable on monocytes and CD11b+HLA-DR+ macrophages but tended to be lowest in the most diseased lung tissue sections. This observation was confirmed by fluorescent microscopy, which showed the expression of CD200R on CD68+ macrophages surrounding TB lung granuloma and found expression levels tended to be lower in macrophages closest to the granuloma core and inversely correlated with lesion size. Antibody blockade of CD200R in a biomimetic 3D granuloma-like tissue culture system led to significantly increased Mtb growth. In addition, Mtb infection in this system reduced gene expression of CD200R. These findings indicate that regulation of myeloid cells via CD200R is likely to play an important part in the immune response to TB and may represent a potential target for novel therapeutic intervention.


Subject(s)
Mycobacterium tuberculosis , Myeloid Cells , Tuberculosis, Pulmonary , Humans , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Orexin Receptors/metabolism , Macrophages/immunology , Macrophages/metabolism , Adult , Female , Male , Antigens, CD/metabolism , Antigens, CD/genetics , Middle Aged , Lung/immunology , Lung/microbiology , Lung/pathology , Lung/metabolism , Biomimetics , Monocytes/immunology , Monocytes/metabolism
18.
J Med Virol ; 96(5): e29678, 2024 May.
Article in English | MEDLINE | ID: mdl-38751128

ABSTRACT

Death due to severe influenza is usually a fatal complication of a dysregulated immune response more than the acute virulence of an infectious agent. Although spleen tyrosine kinase (SYK) as a critical immune signaling molecule and therapeutic target plays roles in airway inflammation and acute lung injury, the role of SYK in influenza virus infection is not clear. Here, we investigated the antiviral and anti-inflammatory effects of SYK inhibitor R406 on influenza infection through a coculture model of human alveolar epithelial (A549) and macrophage (THP-1) cell lines and mouse model. The results showed that R406 treatment increased the viability of A549 and decreased the pathogenicity and mortality of lethal influenza virus in mice with influenza A infection, decreased levels of intracellular signaling molecules under the condition of inflammation during influenza virus infection. Combination therapy with oseltamivir further ameliorated histopathological damage in the lungs of mice and further delayed the initial time to death compared with R406 treatment alone. This study demonstrated that phosphorylation of SYK is involved in the pathogenesis of influenza, and R406 has antiviral and anti-inflammatory effects on the treatment of the disease, which may be realized through multiple pathways, including the already reported SYK/STAT/IFNs-mediated antiviral pathway, as well as TNF-α/SYK- and SYK/Akt-based immunomodulation pathway.


Subject(s)
Anti-Inflammatory Agents , Antiviral Agents , Disease Models, Animal , Orthomyxoviridae Infections , Oxazines , Syk Kinase , Animals , Humans , Syk Kinase/antagonists & inhibitors , Mice , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Oxazines/pharmacology , Oxazines/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Imidazoles/pharmacology , Imidazoles/therapeutic use , Lung/pathology , Lung/virology , Lung/drug effects , Lung/immunology , A549 Cells , Influenza A virus/drug effects , Mice, Inbred BALB C , Oseltamivir/pharmacology , Oseltamivir/therapeutic use , Influenza, Human/drug therapy , Influenza, Human/immunology , THP-1 Cells , Female , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
19.
Bull Exp Biol Med ; 176(5): 680-686, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38733478

ABSTRACT

Morphological, molecular, and biological features of the systemic inflammatory response induced by LPS administration were assessed in adult and old male Wistar rats with high and low resistance to hypoxia. In 6 h after LPS administration, mRNA expression levels of Hif1a, Vegf, Nfkb, and level of IL-1ß protein in old rats were higher than in adult rats regardless of hypoxia tolerance. The morphometric study showed that the number of neutrophils in the interalveolar septa of the lungs was significantly higher in low-resistant adult and old rats 6 h after LPS administration. Thus, in old male Wistar rats, systemic inflammatory response is more pronounced than in adult rats and depends on the initial tolerance to hypoxia, which should be considered when developing new approaches to the therapy of systemic inflammatory response in individuals of different ages.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Hypoxia , Interleukin-1beta , Rats, Wistar , Animals , Male , Rats , Hypoxia/metabolism , Hypoxia/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lipopolysaccharides/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , NF-kappa B/metabolism , NF-kappa B/genetics , Lung/pathology , Lung/metabolism , Lung/drug effects , Lung/immunology , Neutrophils/metabolism , Neutrophils/immunology , Inflammation/metabolism , Inflammation/pathology , Age Factors , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Commun Biol ; 7(1): 584, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755239

ABSTRACT

B cells are important in tuberculosis (TB) immunity, but their role in the human lung is understudied. Here, we characterize B cells from lung tissue and matched blood of patients with TB and found they are decreased in the blood and increased in the lungs, consistent with recruitment to infected tissue, where they are located in granuloma associated lymphoid tissue. Flow cytometry and transcriptomics identify multiple B cell populations in the lung, including those associated with tissue resident memory, germinal centers, antibody secretion, proinflammatory atypical B cells, and regulatory B cells, some of which are expanded in TB disease. Additionally, TB lungs contain high levels of Mtb-reactive antibodies, specifically IgM, which promotes Mtb phagocytosis. Overall, these data reveal the presence of functionally diverse B cell subsets in the lungs of patients with TB and suggest several potential localized roles that may represent a target for interventions to promote immunity or mitigate immunopathology.


Subject(s)
B-Lymphocytes , Humans , B-Lymphocytes/immunology , Lung/immunology , Lung/microbiology , Lung/pathology , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/physiology , Phenotype , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/genetics , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Male , Female , Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...