Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 16.626
Filtrer
1.
Front Immunol ; 15: 1408212, 2024.
Article de Anglais | MEDLINE | ID: mdl-38887303

RÉSUMÉ

Introduction: Varicella zoster virus (VZV) causes varicella and can reactivate as herpes zoster, and both diseases present a significant burden worldwide. However, the mechanisms by which VZV establishes latency in the sensory ganglia and disseminates to these sites remain unclear. Methods: We combined a single-cell sequencing approach and a well-established rhesus macaque experimental model using Simian varicella virus (SVV), which recapitulates the VZV infection in humans, to define the acute immune response to SVV in the lung as well as compare the transcriptome of infected and bystander lung-resident T cells and macrophages. Results and discussion: Our analysis showed a decrease in the frequency of alveolar macrophages concomitant with an increase in that of infiltrating macrophages expressing antiviral genes as well as proliferating T cells, effector CD8 T cells, and T cells expressing granzyme A (GZMA) shortly after infection. Moreover, infected T cells harbored higher numbers of viral transcripts compared to infected macrophages. Furthermore, genes associated with cellular metabolism (glycolysis and oxidative phosphorylation) showed differential expression in infected cells, suggesting adaptations to support viral replication. Overall, these data suggest that SVV infection remodels the transcriptome of bystander and infected lung-resident T cells and macrophages.


Sujet(s)
Poumon , Macaca mulatta , Animaux , Poumon/immunologie , Poumon/virologie , Macrophages alvéolaires/immunologie , Macrophages alvéolaires/virologie , Transcriptome , Lymphocytes T/immunologie , Varicellovirus/physiologie , Varicellovirus/immunologie , Macrophages/immunologie , Macrophages/virologie , Infections à Herpesviridae/immunologie , Infections à Herpesviridae/virologie , Herpèsvirus humain de type 3/immunologie , Herpèsvirus humain de type 3/physiologie , Modèles animaux de maladie humaine , Analyse sur cellule unique
2.
Respir Res ; 25(1): 240, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38867225

RÉSUMÉ

BACKGROUND: Despite the success of antiretroviral therapy (ART), people living with HIV (PLWH) suffer from a high burden of pulmonary diseases, even after accounting for their smoking status. Cytotoxic CD8 T-cells are likely implicated in this phenomenon and may act as a double-edged sword. While being essential in viral infection control, their hyperactivation can also contribute to lung mucosal tissue damage. The effects of HIV and smoking on pulmonary mucosal CD8 T-cell dynamics has been a neglected area of research, which we address herein. METHODS: Bronchoalveolar lavage (BAL) fluid were obtained from ART-treated PLWH (median duration of supressed viral load: 9 years; smokers: n = 14; non-smokers: n = 21) and HIV-uninfected controls (smokers: n = 11; non-smokers: n = 20) without any respiratory symptoms or active infection. Lymphocytes were isolated and CD8 T-cell subsets and homing markers were characterized by multiparametric flow cytometry. RESULTS: Both smoking and HIV infection were independently associated with a significant increase in frequencies of total pulmonary mucosal CD8 T-cell. BAL CD8 T-cells were primarily CD69 + expressing CD103 and/or CD49a, at least one of the two granzymes (GzmA/GzmB), and little Perforin. Higher expression levels of CD103, CD69, and GzmB were observed in smokers versus non-smokers. The ex vivo phenotype of GzmA + and GzmB + cells revealed increased expression of CD103 and CXCR6 in smokers, while PLWH displayed elevated levels of CX3CR1 compared to controls. CONCLUSION: Smoking and HIV could promote cytotoxic CD8 T-cell retention in small airways through different mechanisms. Smoking likely increases recruitment and retention of GzmB + CD8 Trm via CXCR6 and CD103. Heightened CX3CR1 expression could be associated with CD8 non-Trm recruitment from the periphery in PLWH.


Sujet(s)
Infections à VIH , Humains , Mâle , Infections à VIH/traitement médicamenteux , Infections à VIH/immunologie , Femelle , Adulte d'âge moyen , Adulte , Muqueuse respiratoire/immunologie , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/métabolisme , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Lymphocytes T cytotoxiques/métabolisme , Fumer/effets indésirables , Liquide de lavage bronchoalvéolaire/immunologie , Antirétroviraux/usage thérapeutique , Agents antiVIH/usage thérapeutique , Poumon/immunologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme
3.
Biol Open ; 13(6)2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38875000

RÉSUMÉ

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


Sujet(s)
Poumon , microARN , Animaux , microARN/génétique , Souris , Humains , Poumon/virologie , Poumon/immunologie , Poumon/métabolisme , ARN viral/génétique , Interactions hôte-pathogène/génétique , Interactions hôte-pathogène/immunologie , Régulation de l'expression des gènes , Infections de l'appareil respiratoire/immunologie , Infections de l'appareil respiratoire/virologie , Infections de l'appareil respiratoire/génétique , Infections à respirovirus/immunologie
4.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38892470

RÉSUMÉ

The nanosized vesicles secreted from various cell types into the surrounding extracellular space are called extracellular vesicles (EVs). Although mesenchymal stem cell-derived EVs are known to have immunomodulatory effects in asthmatic mice, the role of identified pulmonary genes in the suppression of allergic airway inflammation remains to be elucidated. Moreover, the major genes responsible for immune regulation in allergic airway diseases have not been well documented. This study aims to evaluate the immunomodulatory effects of secretoglobin family 1C member 1 (SCGB1C1) on asthmatic mouse models. C57BL/6 mice were sensitized to ovalbumin (OVA) using intraperitoneal injection and were intranasally challenged with OVA. To evaluate the effect of SCGB1C1 on allergic airway inflammation, 5 µg/50 µL of SCGB1C1 was administrated intranasally before an OVA challenge. We evaluated airway hyperresponsiveness (AHR), total inflammatory cells, eosinophils in the bronchoalveolar lavage fluid (BALF), lung histology, serum immunoglobulin (Ig), the cytokine profiles of BALF and lung-draining lymph nodes (LLN), and the T cell populations in LLNs. The intranasal administration of SCGB1C1 significantly inhibited AHR, the presence of eosinophils in BALF, eosinophilic inflammation, goblet cell hyperplasia in the lung, and serum total and allergen-specific IgE. SCGB1C1 treatment significantly decreased the expression of interleukin (IL)-5 in the BALF and IL-4 in the LLN, but significantly increased the expression of IL-10 and transforming growth factor (TGF)-ß in the BALF. Furthermore, SCGB1C1 treatment notably increased the populations of CD4+CD25+Foxp3+ regulatory T cells (Tregs) in asthmatic mice. The intranasal administration of SCGB1C1 provides a significant reduction in allergic airway inflammation and improvement of lung function through the induction of Treg expansion. Therefore, SCGB1C1 may be the major regulator responsible for suppressing allergic airway inflammation.


Sujet(s)
Asthme , Souris de lignée C57BL , Ovalbumine , Lymphocytes T régulateurs , Animaux , Lymphocytes T régulateurs/immunologie , Souris , Asthme/immunologie , Asthme/métabolisme , Poumon/anatomopathologie , Poumon/immunologie , Poumon/métabolisme , Liquide de lavage bronchoalvéolaire , Cytokines/métabolisme , Modèles animaux de maladie humaine , Immunoglobuline E/sang , Immunoglobuline E/immunologie , Femelle , Inflammation/immunologie , Inflammation/métabolisme , Inflammation/anatomopathologie , Granulocytes éosinophiles/immunologie , Granulocytes éosinophiles/métabolisme
5.
Immun Inflamm Dis ; 12(6): e1307, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38860753

RÉSUMÉ

BACKGROUND: The hygiene hypothesis suggests that early life exposure to helminth infections can reduce hypersensitivity in the immune system. OBJECTIVE: The present study aims to evaluate the effects of Toxocara cati (T. cati) somatic products on allergic airway inflammation. METHODS: Between 2018 and 2020, T. cati adult worms were collected from stray cats in Mashhad, Iran (31 out of 186 cats), and their somatic extract was collected. Thirty BALB/c mice were equally divided into three groups, including the OVA group (sensitized and challenged with ovalbumin), the somatic administered group (received somatic extract along with ovalbumin sensitization), and the PBS group (sensitized and challenged with phosphate buffer saline). Bronchoalveolar lavage (BAL) fluid was collected to assess the number of cells, and lung homogenates were prepared for cytokine analysis. Histopathological analysis of the lungs was performed, and inflammatory cells and mucus were detected. Cytokine levels (IL-4, IL-5, IL-10) were measured using enzyme-linked immunosorbent assay (ELISA), and ovalbumin-specific immunoglobulin E (IgE) levels were determined using a capture ELISA. RESULTS: The somatic group significantly decreased regarding the lung pathological changes, including peribronchiolitis, perivasculitis, and eosinophil influx, compared to the group treated with ovalbumin alone. These changes were accompanied by a decrease in proinflammatory cytokines IL-4 and IL-5 and an increase in the anti-inflammatory cytokine IL-10, indicating a shift toward a more balanced immune response. The number of inflammatory cells in the BAL fluid was also significantly reduced in the somatic group, indicating a decrease in inflammation. CONCLUSION: These preclinical findings suggest that in experimental models, T. cati somatic extract exhibits promising potential as a therapeutic agent for mitigating allergic airway inflammation. Its observed effects on immune response modulation and reduction of inflammatory cell infiltration warrant further investigation in clinical studies to assess its efficacy and safety in human patients.


Sujet(s)
Cytokines , Souris de lignée BALB C , Toxocara , Animaux , Souris , Toxocara/immunologie , Toxocara/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Cytokines/immunologie , Immunoglobuline E/immunologie , Immunoglobuline E/sang , Ovalbumine/immunologie , Poumon/immunologie , Poumon/anatomopathologie , Poumon/parasitologie , Poumon/effets des médicaments et des substances chimiques , Liquide de lavage bronchoalvéolaire/immunologie , Asthme/immunologie , Asthme/traitement médicamenteux , Modèles animaux de maladie humaine , Chats , Femelle , Toxocarose/traitement médicamenteux , Toxocarose/immunologie , Toxocarose/parasitologie
6.
Clin Lab ; 70(6)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38868881

RÉSUMÉ

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.


Sujet(s)
Maladie associée aux immunoglobulines G4 , Immunoglobuline G , Maladies pulmonaires , Tomodensitométrie , Humains , Maladie associée aux immunoglobulines G4/diagnostic , Maladie associée aux immunoglobulines G4/immunologie , Maladies pulmonaires/diagnostic , Maladies pulmonaires/immunologie , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Mâle , Poumon/imagerie diagnostique , Poumon/anatomopathologie , Poumon/immunologie , Adulte d'âge moyen , Biopsie
7.
Nat Commun ; 15(1): 4711, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830841

RÉSUMÉ

The fetal development of organs and functions is vulnerable to perturbation by maternal inflammation which may increase susceptibility to disorders after birth. Because it is not well understood how the placenta and fetus respond to acute lung- inflammation, we characterize the response to maternal pulmonary lipopolysaccharide exposure across 24 h in maternal and fetal organs using multi-omics, imaging and integrative analyses. Unlike maternal organs, which mount strong inflammatory immune responses, the placenta upregulates immuno-modulatory genes, in particular the IL-6 signaling suppressor Socs3. Similarly, we observe no immune response in the fetal liver, which instead displays metabolic changes, including increases in lipids containing docosahexaenoic acid, crucial for fetal brain development. The maternal liver and plasma display similar metabolic alterations, potentially increasing bioavailability of docosahexaenoic acid for the mother and fetus. Thus, our integrated temporal analysis shows that systemic inflammation in the mother leads to a metabolic perturbation in the fetus.


Sujet(s)
Foetus , Lipopolysaccharides , Foie , Poumon , Placenta , Femelle , Grossesse , Placenta/métabolisme , Placenta/immunologie , Animaux , Foetus/immunologie , Foetus/métabolisme , Poumon/immunologie , Poumon/métabolisme , Foie/métabolisme , Foie/immunologie , Acide docosahexaénoïque/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Souris , Inflammation/immunologie , Inflammation/métabolisme , Souris de lignée C57BL , Adaptation physiologique/immunologie , Développement foetal/immunologie , Échange foetomaternel/immunologie , Interleukine-6/métabolisme , Interleukine-6/immunologie
8.
Vet Res ; 55(1): 71, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38822398

RÉSUMÉ

In the wake of the COVID-19 pandemic caused by SARS-CoV-2, questions emerged about the potential effects of Bacillus Calmette-Guérin (BCG) vaccine on the immune response to SARS-CoV-2 infection, including the neurodegenerative diseases it may contribute to. To explore this, an experimental study was carried out in BCG-stimulated and non-stimulated k18-hACE2 mice challenged with SARS-CoV-2. Viral loads in tissues determined by RT-qPCR, histopathology in brain and lungs, immunohistochemical study in brain (IHC) as well as mortality rates, clinical signs and plasma inflammatory and coagulation biomarkers were assessed. Our results showed BCG-SARS-CoV-2 challenged mice presented higher viral loads in the brain and an increased frequency of neuroinvasion, with the greatest differences observed between groups at 3-4 days post-infection (dpi). Histopathological examination showed a higher severity of brain lesions in BCG-SARS-CoV-2 challenged mice, mainly consisting of neuroinflammation, increased glial cell population and neuronal degeneration, from 5 dpi onwards. This group also presented higher interstitial pneumonia and vascular thrombosis in lungs (3-4 dpi), BCG-SARS-CoV-2 mice showed higher values for TNF-α and D-dimer values, while iNOS values were higher in SARS-CoV-2 mice at 3-4 dpi. Results presented in this study indicate that BCG stimulation could have intensified the inflammatory and neurodegenerative lesions promoting virus neuroinvasion and dissemination in this experimental model. Although k18-hACE2 mice show higher hACE2 expression and neurodissemination, this study suggests that, although the benefits of BCG on enhancing heterologous protection against pathogens and tumour cells have been broadly demonstrated, potential adverse outcomes due to the non-specific effects of BCG should be considered.


Sujet(s)
Vaccin BCG , Encéphale , COVID-19 , SARS-CoV-2 , Animaux , Souris , Vaccin BCG/administration et posologie , COVID-19/immunologie , COVID-19/virologie , SARS-CoV-2/physiologie , Encéphale/anatomopathologie , Encéphale/virologie , Charge virale , Poumon/anatomopathologie , Poumon/virologie , Poumon/immunologie , Angiotensin-converting enzyme 2/métabolisme , Souris transgéniques , Femelle
9.
Int J Mol Sci ; 25(11)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38891820

RÉSUMÉ

Chronic obstructive pulmonary disease (COPD), the major leading cause of mortality worldwide, is a progressive and irreversible respiratory condition characterized by peripheral airway and lung parenchymal inflammation, accompanied by fibrosis, emphysema, and airflow limitation, and has multiple etiologies, including genetic variance, air pollution, and repetitive exposure to harmful substances. However, the precise mechanisms underlying the pathogenesis of COPD have not been identified. Recent multiomics-based evidence suggests that the plasticity of alveolar macrophages contributes to the onset and progression of COPD through the coordinated modulation of numerous transcription factors. Therefore, this review focuses on understanding the mechanisms and functions of macrophage polarization that regulate lung homeostasis in COPD. These findings may provide a better insight into the distinct role of macrophages in COPD pathogenesis and perspective for developing novel therapeutic strategies targeting macrophage polarization.


Sujet(s)
Macrophages alvéolaires , Broncho-pneumopathie chronique obstructive , Broncho-pneumopathie chronique obstructive/étiologie , Broncho-pneumopathie chronique obstructive/anatomopathologie , Broncho-pneumopathie chronique obstructive/métabolisme , Broncho-pneumopathie chronique obstructive/génétique , Humains , Macrophages alvéolaires/métabolisme , Macrophages alvéolaires/anatomopathologie , Macrophages alvéolaires/immunologie , Animaux , Activation des macrophages , Macrophages/métabolisme , Macrophages/immunologie , Poumon/anatomopathologie , Poumon/métabolisme , Poumon/immunologie
10.
Int J Mol Sci ; 25(11)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38892358

RÉSUMÉ

Obese patients with asthma present with aggravated symptoms that are also harder to treat. Here, we used a mouse model of allergic asthma sensitised and challenged to house dust mite (HDM) extracts to determine whether high-fat-diet consumption would exacerbate the key features of allergic airway inflammation. C57BL/6 mice were intranasally sensitised and challenged with HDM extracts over a duration of 3 weeks. The impact of high-fat-diet (HFD) vs. normal diet (ND) chow was studied on HDM-induced lung inflammation and inflammatory cell infiltration as well as cytokine production. HFD-fed mice had greater inflammatory cell infiltration around airways and blood vessels, and an overall more severe degree of inflammation than in the ND-fed mice (semiquantitative blinded evaluation). Quantitative assessment of HDM-associated Th2 responses (numbers of lung CD4+ T cells, eosinophils, serum levels of allergen-specific IgE as well as the expression of Th2 cytokines (Il5 and Il13)) did not show significant changes between the HFD and ND groups. Interestingly, the HFD group exhibited a more pronounced neutrophilic infiltration within their lung tissues and an increase in non-Th2 cytokines (Il17, Tnfa, Tgf-b, Il-1b). These findings provide additional evidence that obesity triggered by a high-fat-diet regimen may exacerbate asthma by involving non-Th2 and neutrophilic pathways.


Sujet(s)
Asthme , Cytokines , Alimentation riche en graisse , Modèles animaux de maladie humaine , Souris de lignée C57BL , Obésité , Lymphocytes auxiliaires Th2 , Animaux , Asthme/immunologie , Asthme/étiologie , Asthme/anatomopathologie , Asthme/métabolisme , Obésité/immunologie , Obésité/métabolisme , Souris , Alimentation riche en graisse/effets indésirables , Lymphocytes auxiliaires Th2/immunologie , Lymphocytes auxiliaires Th2/métabolisme , Cytokines/métabolisme , Pyroglyphidae/immunologie , Poumon/anatomopathologie , Poumon/immunologie , Poumon/métabolisme , Inflammation/anatomopathologie , Inflammation/immunologie , Inflammation/métabolisme , Immunoglobuline E/sang , Immunoglobuline E/immunologie , Femelle , Allergènes/immunologie
11.
Immun Inflamm Dis ; 12(6): e1321, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38888451

RÉSUMÉ

BACKGROUND: For decades, studies have demonstrated the anti-inflammatory potential of proteins secreted by helminths in allergies and asthma. Previous studies have demonstrated the immunomodulatory capabilities of Succinate Coenzyme A ligase beta-like protein (SUCLA-ß) derived from Trichinella spiralis, a crucial excretory product of this parasite. OBJECTIVE: To explore the therapeutic potential of SUCLA-ß in alleviating and controlling ovalbumin (OVA)-induced allergic asthma, as well as its influence on host immune modulation. METHODS: In this research, we utilized the rTs-SUCLA-ß protein derived from T. spiralis to investigate its potential in mitigating airway inflammation in a murine model of asthma induced by OVA sensitization/stimulation, both pre- and post-challenge. The treatment's efficacy was assessed by quantifying the extent of inflammation in the lungs. RESULTS: Treatment with rTs-SUCLA-ß demonstrated efficacy in ameliorating OVA-induced airway inflammation, as evidenced by a reduction in eosinophil infiltration, levels of OVA-specific Immunoglobulin E, interferon-γ, interleukin (IL)-9, and IL-17A, along with an elevation in IL-10. The equilibrium between Th17 and Treg cells plays a pivotal role in modulating the abundance of inflammatory cells within the organism, thereby ameliorating inflammation and alleviating symptoms associated with allergic asthma. CONCLUSIONS AND CLINICAL RELEVANCE: Our data revealed that T. spiralis-derived Ts-SUCLA-ß protein may inhibit the allergic airway inflammation by regulating host immune responses.


Sujet(s)
Asthme , Protéines d'helminthes , Ovalbumine , Trichinella spiralis , Trichinella spiralis/immunologie , Animaux , Asthme/immunologie , Asthme/traitement médicamenteux , Souris , Ovalbumine/immunologie , Protéines d'helminthes/immunologie , Protéines d'helminthes/pharmacologie , Souris de lignée BALB C , Modèles animaux de maladie humaine , Femelle , Cytokines/métabolisme , Cytokines/immunologie , Immunoglobuline E/immunologie , Poumon/immunologie , Poumon/parasitologie , Poumon/anatomopathologie , Lymphocytes T régulateurs/immunologie , Hypersensibilité/immunologie , Hypersensibilité/traitement médicamenteux , Cellules Th17/immunologie
12.
PLoS Pathog ; 20(6): e1012222, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38838044

RÉSUMÉ

COVID-19 has affected more than half a billion people worldwide, with more than 6.3 million deaths, but the pathophysiological mechanisms involved in lethal cases and the host determinants that determine the different clinical outcomes are still unclear. In this study, we assessed lung autopsies of 47 COVID-19 patients and examined the inflammatory profiles, viral loads, and inflammasome activation. Additionally, we correlated these factors with the patient's clinical and histopathological conditions. Robust inflammasome activation was detected in the lungs of lethal cases of SARS-CoV-2. Experiments conducted on transgenic mice expressing hACE2 and infected with SARS-CoV-2 showed that Nlrp3-/- mice were protected from disease development and lethality compared to Nlrp3+/+ littermate mice, supporting the involvement of this inflammasome in disease exacerbation. An analysis of gene expression allowed for the classification of COVID-19 patients into two different clusters. Cluster 1 died with higher viral loads and exhibited a reduced inflammatory profile than Cluster 2. Illness time, mechanical ventilation time, pulmonary fibrosis, respiratory functions, histopathological status, thrombosis, viral loads, and inflammasome activation significantly differed between the two clusters. Our data demonstrated two distinct profiles in lethal cases of COVID-19, thus indicating that the balance of viral replication and inflammasome-mediated pulmonary inflammation led to different clinical outcomes. We provide important information to understand clinical variations in severe COVID-19, a process that is critical for decisions between immune-mediated or antiviral-mediated therapies for the treatment of critical cases of COVID-19.


Sujet(s)
COVID-19 , Poumon , SARS-CoV-2 , Charge virale , Réplication virale , COVID-19/virologie , COVID-19/mortalité , COVID-19/immunologie , COVID-19/anatomopathologie , Animaux , Humains , Souris , Femelle , Mâle , Poumon/virologie , Poumon/anatomopathologie , Poumon/immunologie , Adulte d'âge moyen , Inflammasomes/immunologie , Inflammasomes/métabolisme , Sujet âgé , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Souris transgéniques , Pneumopathie infectieuse/virologie , Pneumopathie infectieuse/mortalité , Pneumopathie infectieuse/immunologie , Pneumopathie infectieuse/anatomopathologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Souris knockout , Adulte
13.
Sci Rep ; 14(1): 13524, 2024 06 12.
Article de Anglais | MEDLINE | ID: mdl-38866913

RÉSUMÉ

Myxovirus resistance (Mx) proteins are products of interferon stimulated genes (ISGs) and Mx proteins of different species have been reported to mediate antiviral activity against a number of viruses, including influenza A viruses (IAV). Ferrets are widely considered to represent the 'gold standard' small animal model for studying pathogenesis and immunity to human IAV infections, however little is known regarding the antiviral activity of ferret Mx proteins. Herein, we report induction of ferret (f)Mx1/2 in a ferret lung cell line and in airway tissues from IAV-infected ferrets, noting that fMx1 was induced to higher levels that fMx2 both in vitro and in vivo. Overexpression confirmed cytoplasmic expression of fMx1 as well as its ability to inhibit infection and replication of IAV, noting that this antiviral effect of fMx1was modest when compared to cells overexpressing either human MxA or mouse Mx1. Together, these studies provide the first insights regarding the role of fMx1 in cell innate antiviral immunity to influenza viruses. Understanding similarities and differences in the antiviral activities of human and ferret ISGs provides critical context for evaluating results when studying human IAV infections in the ferret model.


Sujet(s)
Furets , Virus de la grippe A , Protéines de résistance aux myxovirus , Infections à Orthomyxoviridae , Animaux , Protéines de résistance aux myxovirus/génétique , Protéines de résistance aux myxovirus/métabolisme , Virus de la grippe A/immunologie , Humains , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Réplication virale/effets des médicaments et des substances chimiques , Antiviraux/pharmacologie , Lignée cellulaire , Souris , Immunité innée , Poumon/virologie , Poumon/immunologie
14.
PLoS One ; 19(6): e0303334, 2024.
Article de Anglais | MEDLINE | ID: mdl-38848417

RÉSUMÉ

Exercise offers numerous benefits to cancer patients and plays an essential role in postsurgical cancer rehabilitation. However, there is a lack of research examining the effects of exercise after the surgical stress of nephrectomy. To address this gap, we created an animal model that simulated patients who had undergone nephrectomy with or without an exercise intervention. Next, we performed a bioinformatic analysis based on the data generated by the RNA sequencing of the lung tissue sample. An overrepresentation analysis was conducted using two genome databases (Gene Ontology and Kyoto Encyclopedia of Genes and Genomes [KEGG]). A KEGG analysis of the exercise-treated nephrectomy mice revealed enrichment in immune-related pathways, particularly in the NF-κB and B cell-related pathways. The expression of CD79A and IGHD, which are responsible for B cell differentiation and proliferation, was upregulated in the nephrectomy mice. Differential gene expression was categorized as significantly upregulated or downregulated according to nephrectomy and exercise groups. Notably, we identified several gene expression reversals in the nephrectomy groups with exercise that were not found in the nephrectomy without exercise or control groups. Our preliminary results potentially reveal a genetic landscape for the underlying mechanisms of the effects of exercise on our nephrectomy model.


Sujet(s)
Biologie informatique , Poumon , Néphrectomie , Conditionnement physique d'animal , Animaux , Souris , Biologie informatique/méthodes , Poumon/immunologie , Poumon/métabolisme , Mâle , Souris de lignée C57BL , Stress physiologique/immunologie
15.
Int Immunopharmacol ; 136: 112395, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38833845

RÉSUMÉ

Asthma is a long-term disease that causes airways swelling and inflammation and in turn airway narrowing. AdipoRonis an orally active synthetic small molecule that acts as a selective agonist at theadiponectin receptor 1 and 2. The aim of the current study is to delineate the protective effect and the potential underlying mechanism ofadipoRon inairway inflammationinduced byovalbumin (OVA) in comparison withdexamethasone. Adult maleSwiss Albino micewere sensitized to OVA on days 0 and 7, then challenged with OVA on days 14, 15 and 16. AdipoRon was administered orally for 6 days starting from the 11th day till the 16th and 1 h prior to OVA in the challenge days. Obtained results from asthmatic control group showed a significant decrease in serum adiponectin concentration, an increase in inflammatory cell counts inthe bronchoalveolar lavage fluid(BALF), CD68 protein expression, inflammatory cytokine concentration and oxidative stress as well. Administration of adipoRon enhanced antioxidant mechanisms limiting oxidative stress by significantly increasing reduced glutathione (GSH) pulmonary content, decreasing serum lactate dehydrogenase (LDH) together with malondialdehyde (MDA) significant reduction in lung tissue. In addition, it modulated the levels of serum immunoglobulin E (IgE), pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-4, IL-13, nuclear factor kappa B (NF-κB) and the anti-inflammatory one IL-10 improving lung inflammation as revealed by histopathological evaluation. Furthermore, lung tissue expression of nuclear factor erythroid 2-related factor (Nrf2) and 5'AMP-activated protein kinase (AMPK) were significantly increased adipoRon. Notably, results of adipoRon received group were comparable to those of dexamethasone group. In conclusion, our study demonstrates that adipoRon can positively modulate adiponectin expression with activation of AMPK pathway and subsequent improvement in inflammatory and oxidative signaling.


Sujet(s)
AMP-Activated Protein Kinases , Asthme , Modèles animaux de maladie humaine , Ovalbumine , Récepteurs à l'adiponectine , Transduction du signal , Animaux , Asthme/traitement médicamenteux , Asthme/immunologie , Asthme/induit chimiquement , Asthme/métabolisme , Souris , Récepteurs à l'adiponectine/agonistes , Récepteurs à l'adiponectine/métabolisme , Ovalbumine/immunologie , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , AMP-Activated Protein Kinases/métabolisme , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Poumon/immunologie , Cytokines/métabolisme , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Adiponectine , Antiasthmatiques/usage thérapeutique , Antiasthmatiques/pharmacologie , Liquide de lavage bronchoalvéolaire/cytologie , Liquide de lavage bronchoalvéolaire/immunologie , Immunoglobuline E/sang , Humains , Dexaméthasone/usage thérapeutique , Dexaméthasone/pharmacologie , Pipéridines
16.
Nat Commun ; 15(1): 5056, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38871792

RÉSUMÉ

Type 2 innate lymphoid cells (ILC2) initiate early allergic inflammation in the lung, but the factors that promote subsequent resolution of type 2 inflammation and prevent prolonged ILC2 activation are not fully known. Here we show that SLAM-family receptors (SFR) play essential roles in this process. We demonstrate dynamic expression of several SFRs on ILC2s during papain-induced type 2 immunity in mice. SFR deficiency exacerbates ILC2-driven eosinophil infiltration in the lung, and results in a significant increase in IL-13 production by ILC2s exclusively in mediastinal lymph nodes (MLN), leading to increased dendritic cell (DC) and TH2 cell numbers. In MLNs, we observe more frequent interaction between ILC2s and bystander T cells, with T cell-expressed SFRs (especially SLAMF3 and SLAMF5) acting as self-ligands to suppress IL-13 production by ILC2s. Mechanistically, homotypic engagement of SFRs at the interface between ILC2s and T cells delivers inhibitory signaling primarily mediated by SHIP-1. This prevents activation of NF-κB, driven by IL-7 and IL-33, two major drivers of ILC2-mediated type 2 immunity. Thus, our study shows that an ILC2-DC-TH2 regulatory axis may promote the resolution of pulmonary type 2 immune responses, and highlights SLAMF3/SLAMF5 as potential therapeutic targets for ameliorating type 2 immunity.


Sujet(s)
Immunité innée , Inflammation , Poumon , Lymphocytes , Souris de lignée C57BL , Famille des molécules de signalisation de l'activation des lymphocytes , Animaux , Souris , Inflammation/immunologie , Inflammation/métabolisme , Lymphocytes/immunologie , Lymphocytes/métabolisme , Poumon/immunologie , Poumon/anatomopathologie , Famille des molécules de signalisation de l'activation des lymphocytes/métabolisme , Famille des molécules de signalisation de l'activation des lymphocytes/génétique , Papaïne , Lymphocytes auxiliaires Th2/immunologie , Interleukine-13/métabolisme , Interleukine-13/immunologie , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/métabolisme , Interleukine-33/métabolisme , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Souris knockout , Transduction du signal , Facteur de transcription NF-kappa B/métabolisme
17.
Front Immunol ; 15: 1393802, 2024.
Article de Anglais | MEDLINE | ID: mdl-38881896

RÉSUMÉ

Background: Mast cells are critically involved in IgE-mediated diseases, e.g., allergies and asthma. Human mast cells are heterogeneous, and mast cells from different anatomical sites have been shown to respond differently to certain stimuli and drugs. The origin of the mast cells is therefore of importance when setting up a model system, and human lung mast cells are highly relevant cells to study in the context of asthma. We therefore set out to optimize a protocol of IgE-mediated activation of human lung mast cells. Methods: Human lung mast cells were extracted from lung tissue obtained from patients undergoing pulmonary resection by enzyme digestion and mechanical disruption followed by CD117 magnetic-activated cell sorting (MACS) enrichment. Different culturing media and conditions for the IgE-mediated degranulation were tested to obtain an optimized method. Results: IgE crosslinking of human lung mast cells cultured in serum-free media gave a stronger response compared to cells cultured with 10% serum. The addition of stem cell factor (SCF) did not enhance the degranulation. However, when the cells were put in fresh serum-free media 30 minutes prior to the addition of anti-IgE antibodies, the cells responded more vigorously. Maximum degranulation was reached 10 minutes after the addition of anti-IgE. Both CD63 and CD164 were identified as stable markers for the detection of degranulated mast cells over time, while the staining with anti-CD107a and avidin started to decline 10 minutes after activation. The levels of CD203c and CD13 did not change in activated cells and therefore cannot be used as degranulation markers of human lung mast cells. Conclusions: For an optimal degranulation response, human lung mast cells should be cultured and activated in serum-free media. With this method, a very strong and consistent degranulation response with a low donor-to-donor variation is obtained. Therefore, this model is useful for further investigations of IgE-mediated mast cell activation and exploring drugs that target human lung mast cells, for instance, in the context of asthma.


Sujet(s)
Dégranulation cellulaire , Immunoglobuline E , Poumon , Mastocytes , Humains , Mastocytes/immunologie , Mastocytes/métabolisme , Immunoglobuline E/immunologie , Poumon/immunologie , Cellules cultivées , Protéines proto-oncogènes c-kit/immunologie , Protéines proto-oncogènes c-kit/métabolisme , Milieux de culture sans sérum/pharmacologie , Anticorps anti-idiotypiques
18.
Emerg Microbes Infect ; 13(1): 2366359, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38855910

RÉSUMÉ

Tuberculosis (TB) remains a leading cause of mortality among individuals coinfected with HIV, characterized by progressive pulmonary inflammation. Despite TB's hallmark being focal granulomatous lung lesions, our understanding of the histopathological features and regulation of inflammation in HIV & TB coinfection remains incomplete. In this study, we aimed to elucidate these histopathological features through an immunohistochemistry analysis of HIV & TB co-infected and TB patients, revealing marked differences. Notably, HIV & TB granulomas exhibited aggregation of CD68 + macrophage (Mφ), while TB lesions predominantly featured aggregation of CD20+ B cells, highlighting distinct immune responses in coinfection. Spatial transcriptome profiling further elucidated CD68+ Mφ aggregation in HIV & TB, accompanied by activation of IL6 pathway, potentially exacerbating inflammation. Through multiplex immunostaining, we validated two granuloma types in HIV & TB versus three in TB, distinguished by cell architecture. Remarkably, in the two types of HIV & TB granulomas, CD68 + Mφ highly co-expressed IL6R/pSTAT3, contrasting TB granulomas' high IFNGRA/SOCS3 expression, indicating different signaling pathways at play. Thus, activation of IL6 pathway may intensify inflammation in HIV & TB-lungs, while SOCS3-enriched immune microenvironment suppresses IL6-induced over-inflammation in TB. These findings provide crucial insights into HIV & TB granuloma formation, shedding light on potential therapeutic targets, particularly for granulomatous pulmonary under HIV & TB co-infection. Our study emphasizes the importance of a comprehensive understanding of the immunopathogenesis of HIV & TB coinfection and suggests potential avenues for targeting IL6 signaling with SOCS3 activators or anti-IL6R agents to mitigate lung inflammation in HIV & TB coinfected individuals.


Sujet(s)
Co-infection , Granulome , Infections à VIH , Poumon , Macrophages , Récepteurs à l'interleukine-6 , Facteur de transcription STAT-3 , Humains , Co-infection/virologie , Co-infection/immunologie , Co-infection/microbiologie , Infections à VIH/complications , Infections à VIH/immunologie , Macrophages/immunologie , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Granulome/immunologie , Poumon/anatomopathologie , Poumon/immunologie , Récepteurs à l'interleukine-6/métabolisme , Récepteurs à l'interleukine-6/génétique , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Protéine-3 suppressive de la signalisation des cytokine/génétique , Antigènes de différenciation des myélomonocytes/métabolisme , Antigènes de différenciation des myélomonocytes/génétique , Antigènes CD/métabolisme , Antigènes CD/génétique , Transduction du signal , Tuberculose pulmonaire/immunologie , Tuberculose pulmonaire/complications , Mâle , Tuberculose/immunologie , Tuberculose/microbiologie , Tuberculose/complications , Femelle , Adulte , Interleukine-6/métabolisme , Interleukine-6/génétique ,
19.
Parasites Hosts Dis ; 62(2): 243-250, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38835265

RÉSUMÉ

We investigated organ specific Toxocara canis larval migration in mice infected with T. canis larvae. We observed the worm burden and systemic immune responses. Three groups of BALB/c mice (n=5 each) were orally administered 1,000 T. canis 2nd stage larvae to induce larva migrans. Mice were sacrificed at 1, 3, and 5 weeks post-infection. Liver, lung, brain, and eye tissues were collected. Tissue from 2 mice per group was digested for larval count, while the remaining 3 mice underwent histological analysis. Blood hematology and serology were evaluated and compared to that in a control uninfected group (n=5) to assess the immune response. Cytokine levels in bronchoalveolar lavage (BAL) fluid were also analyzed. We found that, 1 week post-infection, the mean parasite load in the liver (72±7.1), brain (31±4.2), lungs (20±5.7), and eyes (2±0) peaked and stayed constant until the 3 weeks. By 5-week post-infection, the worm burden in the liver and lungs significantly decreased to 10±4.2 and 9±5.7, respectively, while they remained relatively stable in the brain and eyes (18±4.2 and 1±0, respectively). Interestingly, ocular larvae resided in all retinal layers, without notable inflammation in outer retina. Mice infected with T. canis exhibited elevated levels of neutrophils, monocytes, eosinophils, and immunoglobulin E. At 5 weeks post-infection, interleukin (IL)-5 and IL-13 levels were elevated in BAL fluid. Whereas IL-4, IL-10, IL-17, and interferon-γ levels in BAL fluid were similar to that in controls. Our findings demonstrate that a small portion of T. canis larvae migrate to the eyes and brain within the first week of infection. Minimal tissue inflammation was observed, probably due to increase of anti-inflammatory cytokines. This study contributes to our understanding of the histological and immunological responses to T. canis infection in mice, which may have implications to further understand human toxocariasis.


Sujet(s)
Encéphale , Cytokines , Larve , Foie , Poumon , Souris de lignée BALB C , Toxocara canis , Toxocarose , Animaux , Toxocara canis/immunologie , Toxocarose/immunologie , Toxocarose/anatomopathologie , Toxocarose/parasitologie , Larve/immunologie , Souris , Cytokines/métabolisme , Poumon/parasitologie , Poumon/immunologie , Poumon/anatomopathologie , Foie/parasitologie , Foie/anatomopathologie , Foie/immunologie , Encéphale/parasitologie , Encéphale/immunologie , Encéphale/anatomopathologie , Liquide de lavage bronchoalvéolaire/immunologie , Liquide de lavage bronchoalvéolaire/parasitologie , Femelle , Charge parasitaire , Oeil/parasitologie , Oeil/immunologie , Oeil/anatomopathologie , Modèles animaux de maladie humaine
20.
Front Immunol ; 15: 1386243, 2024.
Article de Anglais | MEDLINE | ID: mdl-38835757

RÉSUMÉ

Introduction: Current vaccines against COVID-19 administered via parenteral route have limited ability to induce mucosal immunity. There is a need for an effective mucosal vaccine to combat SARS-CoV-2 virus replication in the respiratory mucosa. Moreover, sex differences are known to affect systemic antibody responses against vaccines. However, their role in mucosal cellular responses against a vaccine remains unclear and is underappreciated. Methods: We evaluated the mucosal immunogenicity of a booster vaccine regimen that is recombinant protein-based and administered intranasally in mice to explore sex differences in mucosal humoral and cellular responses. Results: Our results showed that vaccinated mice elicited strong systemic antibody (Ab), nasal, and bronchiole alveolar lavage (BAL) IgA responses, and local T cell immune responses in the lung in a sex-biased manner irrespective of mouse genetic background. Monocytes, alveolar macrophages, and CD103+ resident dendritic cells (DCs) in the lungs are correlated with robust mucosal Ab and T cell responses induced by the mucosal vaccine. Discussion: Our findings provide novel insights into optimizing next-generation booster vaccines against SARS-CoV-2 by inducing spike-specific lung T cell responses, as well as optimizing mucosal immunity for other respiratory infections, and a rationale for considering sex differences in future vaccine research and vaccination practice.


Sujet(s)
Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , Immunité muqueuse , Immunogénicité des vaccins , SARS-CoV-2 , Vaccins sous-unitaires , Animaux , Femelle , Souris , SARS-CoV-2/immunologie , Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , COVID-19/immunologie , COVID-19/virologie , Vaccins sous-unitaires/immunologie , Vaccins sous-unitaires/administration et posologie , Mâle , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Poumon/immunologie , Poumon/virologie , Lymphocytes T/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Souris de lignée C57BL , Administration par voie nasale , Facteurs sexuels , Immunoglobuline A/immunologie , Cellules dendritiques/immunologie , Rappel de vaccin , Immunité humorale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...