Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 7 de 7
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Immunohorizons ; 8(1): 122-135, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-38289252

RÉSUMÉ

Klebsiella pneumoniae (KP) is an extracellular Gram-negative bacterium that causes infections in the lower respiratory and urinary tracts and the bloodstream. STAT1 is a master transcription factor that acts to maintain T cell quiescence under homeostatic conditions. Although STAT1 helps defend against systemic spread of acute KP intrapulmonary infection, whether STAT1 regulation of T cell homeostasis impacts pulmonary host defense during acute bacterial infection and injury is less clear. Using a clinical KP respiratory isolate and a pneumonia mouse model, we found that STAT1 deficiency led to an early neutrophil-dominant transcriptional profile and neutrophil recruitment in the lung preceding widespread bacterial dissemination and lung injury development. Yet, myeloid cell STAT1 was dispensable for control of KP proliferation and dissemination, because myeloid cell-specific STAT1-deficient (LysMCre/WT;Stat1fl/fl) mice showed bacterial burden in the lung, liver, and kidney similar to that of their wild-type littermates. Surprisingly, IL-17-producing CD4+ T cells infiltrated Stat1-/- murine lungs early during KP infection. The increase in Th17 cells in the lung was not due to preexisting immunity against KP and was consistent with circulating rather than tissue-resident CD4+ T cells. However, blocking global IL-17 signaling with anti-IL-17RC administration led to increased proliferation and dissemination of KP, suggesting that IL-17 provided by other innate immune cells is essential in defense against KP. Contrastingly, depletion of CD4+ T cells reduced Stat1-/- murine lung bacterial burden, indicating that early CD4+ T cell activation in the setting of global STAT1 deficiency is pathogenic. Altogether, our findings suggest that STAT1 employs myeloid cell-extrinsic mechanisms to regulate neutrophil responses and provides protection against invasive KP by restricting nonspecific CD4+ T cell activation and immunopathology in the lung.


Sujet(s)
Infections à Klebsiella , Granulocytes neutrophiles , Facteur de transcription STAT-1 , Animaux , Souris , Interleukine-17 , Klebsiella pneumoniae , Poumon/microbiologie , Cellules myéloïdes , Granulocytes neutrophiles/immunologie , Facteur de transcription STAT-1/métabolisme , Infections à Klebsiella/immunologie
2.
J Allergy Clin Immunol ; 151(6): 1513-1524, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-36796454

RÉSUMÉ

BACKGROUND: Inhaled corticosteroids (CSs) are the backbone of asthma treatment, improving quality of life, exacerbation rates, and mortality. Although effective for most, a subset of patients with asthma experience CS-resistant disease despite receiving high-dose medication. OBJECTIVE: We sought to investigate the transcriptomic response of bronchial epithelial cells (BECs) to inhaled CSs. METHODS: Independent component analysis was performed on datasets, detailing the transcriptional response of BECs to CS treatment. The expression of these CS-response components was examined in 2 patient cohorts and investigated in relation to clinical parameters. Supervised learning was used to predict BEC CS responses using peripheral blood gene expression. RESULTS: We identified a signature of CS response that was closely correlated with CS use in patients with asthma. Participants could be separated on the basis of CS-response genes into groups with high and low signature expression. Patients with low expression of CS-response genes, particularly those with a severe asthma diagnosis, showed worse lung function and quality of life. These individuals demonstrated enrichment for T-lymphocyte infiltration in endobronchial brushings. Supervised machine learning identified a 7-gene signature from peripheral blood that reliably identified patients with poor CS-response expression in BECs. CONCLUSIONS: Loss of CS transcriptional responses within bronchial epithelium was related to impaired lung function and poor quality of life, particularly in patients with severe asthma. These individuals were identified using minimally invasive blood sampling, suggesting these findings may enable earlier triage to alternative treatments.


Sujet(s)
Asthme , Qualité de vie , Humains , Asthme/traitement médicamenteux , Asthme/génétique , Asthme/diagnostic , Cellules épithéliales/métabolisme , Hormones corticosurrénaliennes/usage thérapeutique
3.
CHEST Crit Care ; 1(3)2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-38250011

RÉSUMÉ

BACKGROUND: Hospitalized patients with severe COVID-19 follow heterogeneous clinical trajectories, requiring different levels of respiratory support and experiencing diverse clinical outcomes. Differences in host immune responses to SARS-CoV-2 infection may account for the heterogeneous clinical course, but we have limited data on the dynamic evolution of systemic biomarkers and related subphenotypes. Improved understanding of the dynamic transitions of host subphenotypes in COVID-19 may allow for improved patient selection for targeted therapies. RESEARCH QUESTION: We examined the trajectories of host-response profiles in severe COVID-19 and evaluated their prognostic impact on clinical outcomes. STUDY DESIGN AND METHODS: In this prospective observational study, we enrolled 323 inpatients with COVID-19 receiving different levels of baseline respiratory support: (1) low-flow oxygen (37%), (2) noninvasive ventilation (NIV) or high-flow oxygen (HFO; 29%), (3) invasive mechanical ventilation (27%), and (4) extracorporeal membrane oxygenation (7%). We collected plasma samples on enrollment and at days 5 and 10 to measure host-response biomarkers. We classified patients by inflammatory subphenotypes using two validated predictive models. We examined clinical, biomarker, and subphenotype trajectories and outcomes during hospitalization. RESULTS: IL-6, procalcitonin, and angiopoietin 2 persistently were elevated in patients receiving higher levels of respiratory support, whereas soluble receptor of advanced glycation end products (sRAGE) levels displayed the inverse pattern. Patients receiving NIV or HFO at baseline showed the most dynamic clinical trajectory, with 24% eventually requiring intubation and exhibiting worse 60-day mortality than patients receiving invasive mechanical ventilation at baseline (67% vs 35%; P < .0001). sRAGE levels predicted NIV failure and worse 60-day mortality for patients receiving NIV or HFO, whereas IL-6 levels were predictive in all patients regardless of level of support (P < .01). Patients classified to a hyperinflammatory subphenotype at baseline (< 10%) showed worse 60-day survival (P < .0001) and 50% of them remained classified as hyperinflammatory at 5 days after enrollment. INTERPRETATION: Longitudinal study of the systemic host response in COVID-19 revealed substantial and predictive interindividual variability influenced by baseline levels of respiratory support.

4.
J Allergy Clin Immunol ; 146(2): 315-324.e7, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32531372

RÉSUMÉ

BACKGROUND: More than 300 million people carry a diagnosis of asthma, with data to suggest that they are at a higher risk for infection or adverse outcomes from severe acute respiratory syndrome coronavirus 2. Asthma is remarkably heterogeneous, and it is currently unclear how patient-intrinsic factors may relate to coronavirus disease 2019. OBJECTIVE: We sought to identify and characterize subsets of patients with asthma at increased risk for severe acute respiratory syndrome coronavirus 2 infection. METHODS: Participants from 2 large asthma cohorts were stratified using clinically relevant parameters to identify factors related to angiotensin-converting enzyme-2 (ACE2) expression within bronchial epithelium. ACE-2-correlated gene signatures were used to interrogate publicly available databases to identify upstream signaling events and novel therapeutic targets. RESULTS: Stratifying by type 2 inflammatory biomarkers, we identified subjects who demonstrated low peripheral blood eosinophils accompanied by increased expression of the severe acute respiratory syndrome coronavirus 2 receptor ACE2 in bronchial epithelium. Genes highly correlated with ACE2 overlapped with type 1 and 2 IFN signatures, normally induced by viral infections. T-cell recruitment and activation within bronchoalveolar lavage cells of ACE2-high subjects was reciprocally increased. These patients demonstrated characteristics corresponding to risk factors for severe coronavirus disease 2019, including male sex, history of hypertension, low peripheral blood, and elevated bronchoalveolar lavage lymphocytes. CONCLUSIONS: ACE2 expression is linked to upregulation of viral response genes in a subset of type 2-low patients with asthma with characteristics resembling known risk factors for severe coronavirus disease 2019. Therapies targeting the IFN family and T-cell-activating factors may therefore be of benefit in a subset of patients.


Sujet(s)
Asthme/épidémiologie , Asthme/génétique , Infections à coronavirus/épidémiologie , Pandémies , Peptidyl-Dipeptidase A/génétique , Pneumopathie virale/épidémiologie , Récepteurs viraux/génétique , Adolescent , Adulte , Angiotensin-converting enzyme 2 , Asthme/classification , Asthme/immunologie , Betacoronavirus/génétique , Betacoronavirus/immunologie , Marqueurs biologiques/métabolisme , Bronches/immunologie , Bronches/anatomopathologie , Liquide de lavage bronchoalvéolaire/cytologie , Liquide de lavage bronchoalvéolaire/immunologie , COVID-19 , Études de cohortes , Infections à coronavirus/virologie , Granulocytes éosinophiles/immunologie , Granulocytes éosinophiles/anatomopathologie , Femelle , Analyse de profil d'expression de gènes , Humains , Interféron de type I/génétique , Interféron de type I/immunologie , Interféron gamma/génétique , Interféron gamma/immunologie , Mâle , Adulte d'âge moyen , Peptidyl-Dipeptidase A/immunologie , Pneumopathie virale/virologie , Cartographie d'interactions entre protéines , Récepteurs viraux/immunologie , Facteurs de risque , SARS-CoV-2 , Indice de gravité de la maladie , Lymphocytes T/classification , Lymphocytes T/immunologie , Lymphocytes T/anatomopathologie , Transcriptome , États-Unis/épidémiologie
5.
J Exp Med ; 217(7)2020 07 06.
Article de Anglais | MEDLINE | ID: mdl-32399548

RÉSUMÉ

A Th2 immune response is central to allergic airway inflammation, which afflicts millions worldwide. However, the mechanisms that augment GATA3 expression in an antigen-primed developing Th2 cell are not well understood. Here, we describe an unexpected role for Blimp-1, a transcriptional repressor that constrains autoimmunity, as an upstream promoter of GATA3 expression that is critical for Th2 cell development in the lung to inhaled but not systemically delivered allergens but is dispensable for TFH function and IgE production. Mechanistically, Blimp-1 acts through Bcl6, leading to increased GATA3 expression in lung Th2 cells. Surprisingly, the anti-inflammatory cytokine IL-10, but not the pro-inflammatory cytokines IL-6 or IL-21, is required via STAT3 activation to up-regulate Blimp-1 and promote Th2 cell development. These data reveal a hitherto unappreciated role for an IL-10-STAT3-Blimp-1 circuit as an initiator of an inflammatory Th2 response in the lung to allergens. Thus, Blimp-1 in a context-dependent fashion can drive inflammation by promoting rather than terminating effector T cell responses.


Sujet(s)
Allergènes/immunologie , Asthme/immunologie , Poumon/immunologie , Poumon/anatomopathologie , Facteur-1 liant le domaine de régulation positive I/métabolisme , Lymphocytes auxiliaires Th2/immunologie , Animaux , Asthme/complications , Différenciation cellulaire , Facteur de transcription GATA-3/métabolisme , Immunoglobuline E/métabolisme , Inflammation/anatomopathologie , Interleukine-6/métabolisme , Interleukines/métabolisme , Noeuds lymphatiques/métabolisme , Souris de lignée C57BL , Spécificité d'organe , Protéines proto-oncogènes c-bcl-6/métabolisme , Pyroglyphidae/immunologie , Récepteurs à l'interleukine-21/métabolisme , Facteur de transcription STAT-3/métabolisme , Transduction du signal , Lymphocytes T auxiliaires/métabolisme
6.
Blood Adv ; 3(3): 432-445, 2019 02 12.
Article de Anglais | MEDLINE | ID: mdl-30733303

RÉSUMÉ

Thrombocytopenia is associated with worse outcomes in patients with acute respiratory distress syndrome, which is most commonly caused by infection and marked by alveolar-capillary barrier disruption. However, the mechanisms by which platelets protect the lung alveolar-capillary barrier during infectious injury remain unclear. We found that natively thrombocytopenic Mpl -/- mice deficient in the thrombopoietin receptor sustain severe lung injury marked by alveolar barrier disruption and hemorrhagic pneumonia with early mortality following acute intrapulmonary Pseudomonas aeruginosa (PA) infection; barrier disruption was attenuated by platelet reconstitution. Although PA infection was associated with a brisk neutrophil influx, depletion of airspace neutrophils failed to substantially mitigate PA-triggered alveolar barrier disruption in Mpl -/- mice. Rather, PA cell-free supernatant was sufficient to induce lung epithelial cell apoptosis in vitro and in vivo and alveolar barrier disruption in both platelet-depleted mice and Mpl -/- mice in vivo. Cell-free supernatant from PA with genetic deletion of the type 2 secretion system, but not the type 3 secretion system, mitigated lung epithelial cell death in vitro and lung injury in Mpl -/- mice. Moreover, platelet releasates reduced poly (ADP ribose) polymerase cleavage and lung injury in Mpl -/- mice, and boiling of platelet releasates, but not apyrase treatment, abrogated PA supernatant-induced lung epithelial cell cytotoxicity in vitro. These findings indicate that while neutrophil airspace influx does not potentiate infectious lung injury in the thrombocytopenic host, platelets and their factors protect against severe pulmonary complications from pathogen-secreted virulence factors that promote host cell death even in the absence of overt infection.


Sujet(s)
Plaquettes/métabolisme , Lésion pulmonaire/étiologie , Thrombopénie/complications , Animaux , Apoptose , Plaquettes/cytologie , Mort cellulaire , Cellules épithéliales , Lésion pulmonaire/sang , Souris
7.
J Allergy Clin Immunol ; 136(3): 747-756.e4, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-25930197

RÉSUMÉ

BACKGROUND: Single nucleotide polymorphisms in the human gene for the receptor for advanced glycation end-products (RAGE) are associated with an increased incidence of asthma. RAGE is highly expressed in the lung and has been reported to play a vital role in the pathogenesis of murine models of asthma/allergic airway inflammation (AAI) by promoting expression of the type 2 cytokines IL-5 and IL-13. IL-5 and IL-13 are prominently secreted by group 2 innate lymphoid cells (ILC2s), which are stimulated by the proallergic cytokine IL-33. OBJECTIVE: We sought to test the hypothesis that pulmonary RAGE is necessary for allergen-induced ILC2 accumulation in the lung. METHODS: AAI was induced in wild-type and RAGE knockout mice by using IL-33, house dust mite extract, or Alternaria alternata extract. RAGE's lung-specific role in type 2 responses was explored with bone marrow chimeras and induction of gastrointestinal type 2 immune responses. RESULTS: RAGE was found to drive AAI by promoting IL-33 expression in response to allergen and by coordinating the inflammatory response downstream of IL-33. Absence of RAGE impedes pulmonary accumulation of ILC2s in models of AAI. Bone marrow chimera studies suggest that pulmonary parenchymal, but not hematopoietic, RAGE has a central role in promoting AAI. In contrast to the lung, the absence of RAGE does not affect IL-33-induced ILC2 influx in the spleen, type 2 cytokine production in the peritoneum, or mucus hypersecretion in the gastrointestinal tract. CONCLUSIONS: For the first time, this study demonstrates that a parenchymal factor, RAGE, mediates lung-specific accumulation of ILC2s.


Sujet(s)
Asthme/immunologie , Immunité innée , Interleukine-33/immunologie , Poumon/immunologie , Lymphocytes/immunologie , Récepteur spécifique des produits finaux de glycosylation avancée/immunologie , Allergènes/administration et posologie , Allergènes/immunologie , Alternaria/composition chimique , Animaux , Antigènes de Dermatophagoides/administration et posologie , Antigènes de Dermatophagoides/immunologie , Asthme/induit chimiquement , Asthme/génétique , Asthme/anatomopathologie , Moelle osseuse/immunologie , Moelle osseuse/anatomopathologie , Prolifération cellulaire , Tube digestif/immunologie , Tube digestif/anatomopathologie , Régulation de l'expression des gènes , Interleukine-13/génétique , Interleukine-13/immunologie , Interleukine-33/génétique , Interleukine-5/génétique , Interleukine-5/immunologie , Poumon/anatomopathologie , Lymphocytes/anatomopathologie , Souris , Spécificité d'organe , Péritoine/immunologie , Péritoine/anatomopathologie , Pyroglyphidae/composition chimique , Récepteur spécifique des produits finaux de glycosylation avancée/génétique , Transduction du signal , Rate/immunologie , Rate/anatomopathologie , Chimère obtenue par transplantation
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE