Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.176
Filtrer
1.
Trans Am Clin Climatol Assoc ; 134: 94-112, 2024.
Article de Anglais | MEDLINE | ID: mdl-39135597

RÉSUMÉ

Surfactant Protein A (SP-A) is an innate immune modulator produced by the lung with known protective effects against bacteria and viruses. Its role in asthma, an inflammatory lung disease that affects 10% of the world's population, is not entirely known. In this review, we demonstrate that SP-A confers protection against exposure to interleukin-13, a type 2 cytokine integral to eosinophilic asthma, in a mouse model of SP-A deficiency, a house dust mite model of asthma, and in human bronchial epithelial cells from participants with asthma. We also show that small peptides derived from SP-A, such as the major allele of single nucleotide polymorphism (SNP) rs1965708, which includes the carbohydrate recognition domain of SP-A2 at position 223, reduce airway hyperresponsiveness, airway eosinophils, and mucus in a mouse model of asthma. These data suggest that SP-A has beneficial effects relevant to asthma and that an SP-A peptide may have a new therapeutic use in asthma.


Sujet(s)
Asthme , Modèles animaux de maladie humaine , Immunité innée , Protéine A associée au surfactant pulmonaire , Asthme/immunologie , Asthme/traitement médicamenteux , Animaux , Protéine A associée au surfactant pulmonaire/génétique , Protéine A associée au surfactant pulmonaire/métabolisme , Protéine A associée au surfactant pulmonaire/immunologie , Humains , Souris , Polymorphisme de nucléotide simple , Interleukine-13/métabolisme , Interleukine-13/immunologie , Interleukine-13/génétique , Poumon/immunologie , Poumon/métabolisme , Antiasthmatiques/pharmacologie , Antiasthmatiques/usage thérapeutique , Pyroglyphidae/immunologie
2.
Clin Transl Sci ; 17(8): e13899, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39080841

RÉSUMÉ

Allergic disease prevalence has increased globally with the subset of type 2 inflammatory diseases playing a substantial role. Type 2 inflammatory diseases may differ in clinical presentation, but they exhibit shared pathophysiology that is targeted by the unique pharmacology of dupilumab. Dupilumab binds to the interleukin (IL)-4 receptor alpha subunit (IL-4Rα) that blocks IL-4 and IL-13 signaling, two key drivers of type 2 inflammation. Herein, we review the mechanism of action and pharmacology of dupilumab, and the clinical evidence that led to the regulatory approvals of dupilumab for the treatment of numerous type 2 inflammatory diseases: atopic dermatitis, asthma, chronic rhinosinusitis with nasal polyposis, eosinophilic esophagitis, and prurigo nodularis.


Sujet(s)
Anticorps monoclonaux humanisés , Eczéma atopique , Interleukine-13 , Sous-unité alpha du récepteur à l'interleukine-4 , , Humains , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/pharmacologie , Sous-unité alpha du récepteur à l'interleukine-4/antagonistes et inhibiteurs , Sous-unité alpha du récepteur à l'interleukine-4/métabolisme , Eczéma atopique/traitement médicamenteux , Eczéma atopique/immunologie , Interleukine-13/antagonistes et inhibiteurs , Interleukine-13/métabolisme , Interleukine-13/immunologie , Interleukine-4/antagonistes et inhibiteurs , Interleukine-4/métabolisme , Asthme/traitement médicamenteux , Asthme/immunologie , Oesophagite à éosinophiles/traitement médicamenteux , Oesophagite à éosinophiles/immunologie , Transduction du signal/effets des médicaments et des substances chimiques , Polypes du nez/traitement médicamenteux , Polypes du nez/immunologie , Prurigo/traitement médicamenteux , Science biomédicale translationnelle , Sinusite/traitement médicamenteux , Sinusite/immunologie
3.
J Immunol ; 213(6): 831-842, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39082935

RÉSUMÉ

Calcitriol, the bioactive form of vitamin D, exerts its biological functions by binding to its cognate receptor, the vitamin D receptor (VDR). The indicators of the severity of allergies and asthma have been linked to low vitamin D levels. However, the role of calcitriol in regulating IL-4 and IL-13, two cytokines pivotal to allergic inflammation, remained unclear. Our study observed diminished IL-4 and IL-13 secretion in murine and human Th2 cells treated with calcitriol. In murine Th2 cells, Gata3 expression was attenuated by calcitriol. However, the expression of the transcriptional repressor Gfi1, too, was attenuated in the presence of calcitriol. Ectopic expression of either Gfi1 or VDR impaired the secretion of IL-13 in Th2 cells. In murine Th2 cells, VDR interacted with Gata3 but not Gfi1. Gfi1 significantly impaired Il13 promoter activation, which calcitriol failed to restore. Conversely, calcitriol augmented Gfi1 recruitment to the Il13 promoter. Ecr, a conserved region between these two genes, which enhanced the transactivation of Il4 and Il13 promoters, is essential for calcitriol-mediated suppression of both the genes. Calcitriol augmented the recruitment of VDR to the Il13 promoter and Ecr regions. Gata3 recruitment was significantly impaired at the Il13 and Ecr loci in the presence of calcitriol but increased at the Il4 promoter. Furthermore, the recruitment of the histone deacetylase HDAC1 was universally increased at the promoters of Il4, Il13, and Ecr when calcitriol was present. Together, our data clearly elucidate that calcitriol modulates VDR, Gata3, and Gfi1 to suppress IL-4 and IL-13 production in Th2 cells.


Sujet(s)
Calcitriol , Facteur de transcription GATA-3 , Interleukine-13 , Interleukine-4 , Récepteur calcitriol , Lymphocytes auxiliaires Th2 , Facteur de transcription GATA-3/métabolisme , Facteur de transcription GATA-3/génétique , Récepteur calcitriol/métabolisme , Récepteur calcitriol/génétique , Calcitriol/pharmacologie , Animaux , Interleukine-4/métabolisme , Interleukine-4/immunologie , Interleukine-13/métabolisme , Interleukine-13/immunologie , Souris , Lymphocytes auxiliaires Th2/immunologie , Humains , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique
4.
Immunotherapy ; 16(13): 845-852, 2024.
Article de Anglais | MEDLINE | ID: mdl-39073081

RÉSUMÉ

Dupilumab has been approved to treat a variety of atopic disorders and was the first US FDA-approved medication for the treatment of eosinophilic esophagitis (EoE), initially approved in May 2022, with expansion in use to patients as young as 1 year of age weighing at least 15 kg in January 2024. It is a fully human monoclonal antibody that inhibits both IL-4 and IL-13 signaling, suppressing TH2-mediated proinflammatory cytokines, chemokines and IgE implicated in EoE pathogenesis. Phase II and III trials in EoE have demonstrated histologic, endoscopic and symptomatic improvement in disease activity with an overall favorable safety profile. This article will review the available clinical trial data and real-world efficacy of dupilumab in EoE.


Dupilumab is a biologic medication used for the treatment of eosinophilic esophagitis. Clinical trials have shown that this medication is effective in treating both inflammation in the esophagus and symptoms associated with eosinophilic esophagitis in a high proportion of patients. Dupilumab was well tolerated by the majority of clinical trial patients, though side effects such as injection site redness and swelling have been reported. More serious side effects are overall rare.


Sujet(s)
Anticorps monoclonaux humanisés , Oesophagite à éosinophiles , Interleukine-13 , Oesophagite à éosinophiles/traitement médicamenteux , Oesophagite à éosinophiles/immunologie , Humains , Anticorps monoclonaux humanisés/usage thérapeutique , Interleukine-13/antagonistes et inhibiteurs , Interleukine-13/métabolisme , Interleukine-13/immunologie , Interleukine-4/métabolisme , Interleukine-4/antagonistes et inhibiteurs , Essais cliniques comme sujet , Résultat thérapeutique , Lymphocytes auxiliaires Th2/immunologie
5.
Nat Commun ; 15(1): 5949, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009587

RÉSUMÉ

Bullous pemphigoid (BP) is a type 2 inflammation- and immunity-driven skin disease, yet a comprehensive understanding of the immune landscape, particularly immune-stromal crosstalk in BP, remains elusive. Herein, using single-cell RNA sequencing (scRNA-seq) and in vitro functional analyzes, we pinpoint Th2 cells, dendritic cells (DCs), and fibroblasts as crucial cell populations. The IL13-IL13RA1 ligand-receptor pair is identified as the most significant mediator of immune-stromal crosstalk in BP. Notably, fibroblasts and DCs expressing IL13RA1 respond to IL13-secreting Th2 cells, thereby amplifying Th2 cell-mediated cascade responses, which occurs through the specific upregulation of PLA2G2A in fibroblasts and CCL17 in myeloid cells, creating a positive feedback loop integral to immune-stromal crosstalk. Furthermore, PLA2G2A and CCL17 contribute to an increased titer of pathogenic anti-BP180-NC16A autoantibodies in BP patients. Our work provides a comprehensive insight into BP pathogenesis and shows a mechanism governing immune-stromal interactions, providing potential avenues for future therapeutic research.


Sujet(s)
Chimiokine CCL17 , Cellules dendritiques , Fibroblastes , Pemphigoïde bulleuse , Analyse sur cellule unique , Lymphocytes auxiliaires Th2 , Humains , Pemphigoïde bulleuse/immunologie , Pemphigoïde bulleuse/génétique , Analyse sur cellule unique/méthodes , Fibroblastes/métabolisme , Fibroblastes/immunologie , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Chimiokine CCL17/génétique , Chimiokine CCL17/métabolisme , Lymphocytes auxiliaires Th2/immunologie , Autoanticorps/immunologie , Transcriptome , Interleukine-13/métabolisme , Interleukine-13/génétique , Interleukine-13/immunologie , Collagènes non fibrillaires/immunologie , Collagènes non fibrillaires/génétique , Collagènes non fibrillaires/métabolisme , Inflammation/immunologie , Inflammation/génétique , Inflammation/métabolisme , Analyse de profil d'expression de gènes/méthodes , Mâle , Femelle , Autoantigènes/immunologie , Autoantigènes/métabolisme , Autoantigènes/génétique , , Cellules myéloïdes/métabolisme , Cellules myéloïdes/immunologie , Cellules stromales/métabolisme , Cellules stromales/immunologie
6.
Pediatr Allergy Immunol ; 35(6): e14181, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38934228

RÉSUMÉ

Atopic dermatitis (AD) is still a demanding challenge in clinical practice. Type 2 inflammation is the most common inflammatory pathway in children and adolescents with AD. Anti-inflammatory drugs, mainly corticosteroids (CS) and immunomodulant agents are the primary therapeutic approach to dampening type 2 inflammation. However, AD patients may require long-term high CS doses or drug combinations with possibly significant adverse effects to achieve and maintain disease control. In this regard, the advent of biologics constituted a breakthrough in managing this condition. Dupilumab is a monoclonal antibody directed against the IL-4 receptor α-subunit (IL-4Rα), antagonizing both IL-4 and IL-13 and is approved for pediatric severe AD. This review presents and discusses the most recent published studies on dupilumab in children and adolescents with AD. There is convincing evidence that dupilumab is safe and effective in managing AD. It can reduce skin lesions and associated itching, reduce the need for additional medications, and improve disease control and quality of life. However, a thorough diagnostic pathway is mandatory, especially considering the different AD phenotypes. The ideal eligible candidate is a child or adolescent with AD requiring systemic treatment because of severe clinical manifestations and impaired quality of life.


Sujet(s)
Anticorps monoclonaux humanisés , Eczéma atopique , Humains , Eczéma atopique/traitement médicamenteux , Anticorps monoclonaux humanisés/usage thérapeutique , Adolescent , Enfant , Sous-unité alpha du récepteur à l'interleukine-4/antagonistes et inhibiteurs , Sous-unité alpha du récepteur à l'interleukine-4/immunologie , Indice de gravité de la maladie , Interleukine-4/antagonistes et inhibiteurs , Interleukine-4/immunologie , Qualité de vie , Interleukine-13/antagonistes et inhibiteurs , Interleukine-13/immunologie , Résultat thérapeutique
7.
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
9.
Biomed Res ; 45(3): 115-123, 2024.
Article de Anglais | MEDLINE | ID: mdl-38839354

RÉSUMÉ

Mixed lymphocyte culture under the blockade of CD80/CD86-CD28 co-stimulation induces anergic (completely hyporesponsive) T cells with immune suppressive function (inducible suppressing T cells: iTS cells). Previously, iTS cell therapy has demonstrated outstanding benefits in clinical trials for organ transplantation. Here, we examined whether peptide antigen-specific iTS cells are inducible. DO 11.10 iTS cells were obtained from splenocytes of BALB/c DO 11.10 mice by stimulation with OVA peptide and antagonistic anti-CD80/CD86 mAbs. When DO 11.10 iTS or Foxp3- DO 11.10 iTS cells were stimulated with OVA, these cells produced IL-13, but not IL-4. DO 11.10 iTS cells decreased IL-4 and increased IL-13 production from OVA-stimulated naïve DO 11.10 splenocytes. When Foxp3+ DO 11.10 iTS cells were prepared, these cells significantly inhibited the production of IL-4 and IL-13 compared with freshly isolated Foxp3+ DO 11.10 T cells. Moreover, an increase in the population expressing OX40, ICOS, and 4-1BB suggested activation of Foxp3+ DO 11.10 iTS cells. Thus, blockade of CD80/CD86-CD28 co-stimulation during peptide antigen stimulation augments the inhibitory function of Foxp3+ regulatory T cells, and does not induce anergic Foxp3- conventional T cells. Peptide-specific Foxp3+ regulatory iTS cells could be useful for the treatment of allergic and autoimmune diseases without adverse effects.


Sujet(s)
Antigène CD80 , Antigène CD86 , Antigène CD28 , Lymphocytes T régulateurs , Animaux , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Antigène CD28/immunologie , Antigène CD28/métabolisme , Souris , Antigène CD80/métabolisme , Antigène CD80/immunologie , Antigène CD86/métabolisme , Antigène CD86/immunologie , Souris de lignée BALB C , Facteurs de transcription Forkhead/métabolisme , Peptides/pharmacologie , Peptides/immunologie , Activation des lymphocytes/immunologie , Interleukine-4/métabolisme , Interleukine-4/immunologie , Interleukine-13/métabolisme , Interleukine-13/immunologie , Ovalbumine/immunologie , Rate/immunologie , Rate/cytologie , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/immunologie
10.
Clin Transl Sci ; 17(6): e13864, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38924698

RÉSUMÉ

Lunsekimig is a novel, bispecific NANOBODY® molecule that inhibits both thymic stromal lymphopoietin (TSLP) and interleukin (IL)-13, two key mediators of asthma pathophysiology. In this first-in-human study, we evaluated the safety, tolerability, pharmacokinetics (PK), pharmacodynamics (PD), and immunogenicity of lunsekimig in healthy adult participants. Participants received single ascending doses (SAD) of lunsekimig (10-400 mg intravenous [IV] or 400 mg subcutaneous [SC]) (SAD part) or multiple ascending doses (MAD part) of lunsekimig (100 or 200 mg, every 2 weeks [Q2W] for three SC doses), or placebo. Overall, 48 participants were randomized 3:1 in the SAD part and 4:1 in the MAD part for lunsekimig or placebo. The primary endpoint was safety and tolerability. The secondary endpoints included PK, antidrug antibodies (ADAs) and total target measurement. Lunsekimig was well tolerated and common treatment-emergent adverse events were COVID-19, nasopharyngitis, injection site reactions, and headache. Lunsekimig showed dose-proportional increases in exposure and linear elimination. Mean t1/2z of lunsekimig was around 10 days across all IV and SC doses of the SAD and MAD parts of the study. Increases in the serum concentration of total TSLP and IL-13 for lunsekimig versus placebo indicated target engagement. ADA of low titers were detected in four (11.1%) participants who received lunsekimig in the SAD, and seven (43.8%) in the MAD. In conclusion, lunsekimig was well tolerated in healthy participants with a linear PK profile up to single 400 mg IV and SC dose and multiple doses of 100 and 200 mg SC Q2W, with low immunogenicity.


Sujet(s)
Cytokines , Volontaires sains , Interleukine-13 , Anticorps à domaine unique , Lymphopoïétine stromale thymique , Humains , Adulte , Mâle , Femelle , Interleukine-13/antagonistes et inhibiteurs , Interleukine-13/immunologie , Anticorps à domaine unique/administration et posologie , Anticorps à domaine unique/immunologie , Anticorps à domaine unique/effets indésirables , Adulte d'âge moyen , Cytokines/immunologie , Cytokines/sang , Jeune adulte , Injections sous-cutanées , Méthode en double aveugle , Relation dose-effet des médicaments , Calendrier d'administration des médicaments
11.
Anaerobe ; 88: 102860, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38701912

RÉSUMÉ

OBJECTIVES: Clostridioides difficile infection (CDI) is the leading hospital-acquired infection in North America. We have previously discovered that antibiotic disruption of the gut microbiota decreases intestinal IL-33 and IL-25 and increases susceptibility to CDI. We further found that IL-33 promotes protection through type 2 Innate Lymphoid Cells (ILC2s), which produce IL-13. However, the contribution of IL-13 to disease has never been explored. METHODS: We used a validated model of CDI in mice, in which we neutralized via blocking antibodies, or administered recombinant protein, IL-13 to assess the role of this cytokine during infection using weight and clinical scores. Fluorescent activated cell sorting (FACS) was used to characterize myeloid cell population changes in response to IL-13 manipulation. RESULTS: We found that administration of IL-13 protected, and anti-IL-13 exacerbated CDI. Additionally, we observed alterations to the monocyte/macrophage cells following neutralization of IL-13 as early as day three post infection. We also observed elevated accumulation of myeloid cells by day four post-infection following IL-13 neutralization. Neutralization of the decoy receptor, IL-13Rα2, resulted in protection from disease, likely through increased available endogenous IL-13. CONCLUSIONS: Our data highlight the protective role of IL-13 in protecting from more severe CDI and the association of poor responses with a dysregulated monocyte-macrophage compartment. These results increase our understanding of type 2 immunity in CDI and may have implications for treating disease in patients.


Sujet(s)
Clostridioides difficile , Infections à Clostridium , Modèles animaux de maladie humaine , Interleukine-13 , Animaux , Souris , Clostridioides difficile/immunologie , Infections à Clostridium/prévention et contrôle , Infections à Clostridium/immunologie , Infections à Clostridium/microbiologie , Colite/immunologie , Colite/prévention et contrôle , Colite/microbiologie , Interleukine-13/métabolisme , Interleukine-13/immunologie , Souris de lignée C57BL , Mâle
12.
Nat Immunol ; 25(6): 1059-1072, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38802511

RÉSUMÉ

Asthma, the most prevalent respiratory disease, affects more than 300 million people and causes more than 250,000 deaths annually. Type 2-high asthma is characterized by interleukin (IL)-5-driven eosinophilia, along with airway inflammation and remodeling caused by IL-4 and IL-13. Here we utilize IL-5 as the targeting domain and deplete BCOR and ZC3H12A to engineer long-lived chimeric antigen receptor (CAR) T cells that can eradicate eosinophils. We call these cells immortal-like and functional IL-5 CAR T cells (5TIF) cells. 5TIF cells were further modified to secrete an IL-4 mutein that blocks IL-4 and IL-13 signaling, designated as 5TIF4 cells. In asthma models, a single infusion of 5TIF4 cells in fully immunocompetent mice, without any conditioning regimen, led to sustained repression of lung inflammation and alleviation of asthmatic symptoms. These data show that asthma, a common chronic disease, can be pushed into long-term remission with a single dose of long-lived CAR T cells.


Sujet(s)
Asthme , Récepteurs chimériques pour l'antigène , Animaux , Asthme/immunologie , Asthme/thérapie , Souris , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Immunothérapie adoptive/méthodes , Lymphocytes T/immunologie , Interleukine-5/immunologie , Interleukine-5/métabolisme , Modèles animaux de maladie humaine , Humains , Interleukine-4/immunologie , Interleukine-4/métabolisme , Souris de lignée C57BL , Granulocytes éosinophiles/immunologie , Femelle , Interleukine-13/métabolisme , Interleukine-13/immunologie
13.
Front Immunol ; 15: 1356298, 2024.
Article de Anglais | MEDLINE | ID: mdl-38690264

RÉSUMÉ

Chronic rhinosinusitis with nasal polyps (CRSwNP) is predominantly a type 2 inflammatory disease associated with type 2 (T2) cell responses and epithelial barrier, mucociliary, and olfactory dysfunction. The inflammatory cytokines interleukin (IL)-4, IL-13, and IL-5 are key mediators driving and perpetuating type 2 inflammation. The inflammatory responses driven by these cytokines include the recruitment and activation of eosinophils, basophils, mast cells, goblet cells, M2 macrophages, and B cells. The activation of these immune cells results in a range of pathologic effects including immunoglobulin E production, an increase in the number of smooth muscle cells within the nasal mucosa and a reduction in their contractility, increased deposition of fibrinogen, mucus hyperproduction, and local edema. The cytokine-driven structural changes include nasal polyp formation and nasal epithelial tissue remodeling, which perpetuate barrier dysfunction. Type 2 inflammation may also alter the availability or function of olfactory sensory neurons contributing to loss of sense of smell. Targeting these key cytokine pathways has emerged as an effective approach for the treatment of type 2 inflammatory airway diseases, and a number of biologic agents are now available or in development for CRSwNP. In this review, we provide an overview of the inflammatory pathways involved in CRSwNP and describe how targeting key drivers of type 2 inflammation is an effective therapeutic option for patients.


Sujet(s)
Interleukine-13 , Interleukine-4 , Polypes du nez , , Animaux , Humains , Maladie chronique , Inflammation/immunologie , Inflammation/métabolisme , Interleukine-13/métabolisme , Interleukine-13/immunologie , Interleukine-4/métabolisme , Interleukine-4/immunologie , Muqueuse nasale/immunologie , Muqueuse nasale/métabolisme , Muqueuse nasale/anatomopathologie , Polypes du nez/immunologie , Polypes du nez/métabolisme , /immunologie , /métabolisme , Transduction du signal
14.
Immunity ; 57(6): 1260-1273.e7, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38744292

RÉSUMÉ

Upon parasitic helminth infection, activated intestinal tuft cells secrete interleukin-25 (IL-25), which initiates a type 2 immune response during which lamina propria type 2 innate lymphoid cells (ILC2s) produce IL-13. This causes epithelial remodeling, including tuft cell hyperplasia, the function of which is unknown. We identified a cholinergic effector function of tuft cells, which are the only epithelial cells that expressed choline acetyltransferase (ChAT). During parasite infection, mice with epithelial-specific deletion of ChAT had increased worm burden, fitness, and fecal egg counts, even though type 2 immune responses were comparable. Mechanistically, IL-13-amplified tuft cells release acetylcholine (ACh) into the gut lumen. Finally, we demonstrated a direct effect of ACh on worms, which reduced their fecundity via helminth-expressed muscarinic ACh receptors. Thus, tuft cells are sentinels in naive mice, and their amplification upon helminth infection provides an additional type 2 immune response effector function.


Sujet(s)
Acétylcholine , Muqueuse intestinale , Animaux , Acétylcholine/métabolisme , Souris , Muqueuse intestinale/immunologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/parasitologie , Choline O-acetyltransferase/métabolisme , Interleukine-13/métabolisme , Interleukine-13/immunologie , Souris knockout , Souris de lignée C57BL , Helminthiase/immunologie , Helminthiase/parasitologie , Cellules épithéliales/immunologie , Cellules épithéliales/métabolisme , Immunité innée , Nematospiroides dubius/immunologie ,
16.
J Food Sci ; 89(5): 3037-3047, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38563099

RÉSUMÉ

Bovine casein is a major allergen present in cow milk to induce anaphylaxis. In this study, the potential allergenicity of enzymatically hydrolyzed casein (HC) was evaluated based on in vitro and in vivo. The results showed that Alcalase and Protamex treatment (AT, PT) reduced the potential allergenicity of CN, with the greatest reductions of 68.25% and 50.75%, respectively. In addition, in vivo results showed that HC effectively alleviated allergic response symptoms of Balb/c mice; a significant tendency toward decreased serum IgG1 and mast cell tryptase levels was observed, accompanied by a decrease of Th2-associated IL-4, IL-5, and IL-13 and an increase of IFN-γ levels in spleen. Moreover, the inflammation of the lung, jejunum, and ileum was remarkably ameliorated. The findings indicated that HC induced a shift toward Th1 response and maintained the Th1/Th2 immune balance. Importantly, our results provide the basis for the production of hypoallergenic dairy products.


Sujet(s)
Allergènes , Caséines , Souris de lignée BALB C , Lymphocytes auxiliaires Th2 , Animaux , Souris , Caséines/immunologie , Allergènes/immunologie , Femelle , Lymphocytes auxiliaires Th2/immunologie , Hydrolyse , Immunoglobuline G/sang , Modèles animaux de maladie humaine , Bovins , Rate/immunologie , Hypersensibilité au lait/immunologie , Interféron gamma/métabolisme , Lymphocytes auxiliaires Th1/immunologie , Interleukine-4/métabolisme , Tryptases/métabolisme , Cytokines/métabolisme , Jéjunum/immunologie , Lait/immunologie , Lait/composition chimique , Interleukine-13/immunologie , Interleukine-13/métabolisme , Anaphylaxie/immunologie , Anaphylaxie/induit chimiquement , Anaphylaxie/prévention et contrôle , Interleukine-5/immunologie
17.
J Interferon Cytokine Res ; 44(9): 399-407, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38516928

RÉSUMÉ

Interleukin (IL)-4 and IL-13 are the main effectors of innate lymphoid cells (ILC2) of the type 2 innate immune response, which can carry out specific signal transmission between multiple cells in the tumor immune microenvironment. IL-4 and IL-13 mediate signal transduction and regulate cellular functions in a variety of solid tumors through their shared receptor chain, the transmembrane heterodimer interleukin-4 receptor alpha/interleukin-13 receptor alpha-1 (type II IL-4 receptor). IL-4, IL-13, and their receptors can induce the formation of a variety of malignant tumors and play an important role in their progression, growth, and tumor immunity. In order to explore possible targets for lung cancer prediction and treatment, this review summarizes the characteristics and signal transduction pathways of IL-4 and IL-13, and their respective receptors, and discusses in depth their possible role in the occurrence and development of lung cancer.


Sujet(s)
Interleukine-13 , Interleukine-4 , Tumeurs du poumon , Humains , Tumeurs du poumon/immunologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Interleukine-13/métabolisme , Interleukine-13/immunologie , Interleukine-4/métabolisme , Interleukine-4/immunologie , Animaux , Transduction du signal/immunologie , Récepteurs à l'interleukine-4/métabolisme , Récepteurs à l'interleukine-4/immunologie , Récepteurs à l'interleukine-13/métabolisme , Récepteurs à l'interleukine-13/immunologie , Microenvironnement tumoral/immunologie
18.
J Allergy Clin Immunol ; 153(5): 1355-1368, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38310974

RÉSUMÉ

BACKGROUND: Eosinophilic esophagitis (EoE) is an increasingly common inflammatory condition of the esophagus; however, the underlying immunologic mechanisms remain poorly understood. The epithelium-derived cytokine IL-33 is associated with type 2 immune responses and elevated in esophageal biopsy specimens from patients with EoE. OBJECTIVE: We hypothesized that overexpression of IL-33 by the esophageal epithelium would promote the immunopathology of EoE. METHODS: We evaluated the functional consequences of esophageal epithelial overexpression of a secreted and active form of IL-33 in a novel transgenic mouse, EoE33. EoE33 mice were analyzed for clinical and immunologic phenotypes. Esophageal contractility was assessed. Epithelial cytokine responses were analyzed in three-dimensional organoids. EoE33 phenotypes were further characterized in ST2-/-, eosinophil-deficient, and IL-13-/- mice. Finally, EoE33 mice were treated with dexamethasone. RESULTS: EoE33 mice displayed ST2-dependent, EoE-like pathology and failed to thrive. Esophageal tissue remodeling and inflammation included basal zone hyperplasia, eosinophilia, mast cells, and TH2 cells. Marked increases in levels of type 2 cytokines, including IL-13, and molecules associated with immune responses and tissue remodeling were observed. Esophageal organoids suggested reactive epithelial changes. Genetic deletion of IL-13 in EoE33 mice abrogated pathologic changes in vivo. EoE33 mice were responsive to steroids. CONCLUSIONS: IL-33 overexpression by the esophageal epithelium generated immunopathology and clinical phenotypes resembling human EoE. IL-33 may play a pivotal role in the etiology of EoE by activating the IL-13 pathway. EoE33 mice are a robust experimental platform for mechanistic investigation and translational discovery.


Sujet(s)
Oesophagite à éosinophiles , Interleukine-13 , Interleukine-33 , Animaux , Humains , Souris , Modèles animaux de maladie humaine , Oesophagite à éosinophiles/immunologie , Oesophagite à éosinophiles/génétique , Oesophagite à éosinophiles/anatomopathologie , Granulocytes éosinophiles/immunologie , Muqueuse oesophagienne/anatomopathologie , Muqueuse oesophagienne/immunologie , Oesophage/anatomopathologie , Oesophage/immunologie , Protéine-1 analogue au récepteur de l'interleukin-1/génétique , Protéine-1 analogue au récepteur de l'interleukin-1/métabolisme , Interleukine-13/génétique , Interleukine-13/immunologie , Interleukine-13/métabolisme , Interleukine-33/génétique , Interleukine-33/immunologie , Interleukine-33/métabolisme , Souris de lignée C57BL , Souris knockout , Souris transgéniques
19.
J Asthma ; 61(7): 762-765, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38152869

RÉSUMÉ

INTRODUCTION: Interleukin (IL)-4 and IL-13 are considered key drivers of type 2 inflammatory diseases. Dupilumab is a fully human monoclonal antibody that blocks the shared receptor component for IL-4 and IL-13, thus inhibiting signaling of both cytokines. CASE STUDY: We report a case of a patient with uncontrolled severe asthma and other T2 inflammatory diseases (atopic dermatitis, chronic rhinosinusitis with nasal polyposis and eosinophilic esophagitis) treated with dupilumab. RESULTS: After one year of treatment, dupilumab improved asthma control together with lung function parameters and airway inflammation. Additionally, a positive impact on quality of life (QoL), evaluated by validated questionnaires, across all the diseases was observed. CONCLUSION: In this case report, a positive and objectively measurable of global improvement on QoL across all four T2 comorbidities was observed after treatment with dupilumab, demonstrating the important role of IL-4 and IL-13 and the existence of a unifying pathological mechanism in T2 diseases.


Sujet(s)
Anticorps monoclonaux humanisés , Asthme , Eczéma atopique , Oesophagite à éosinophiles , Polypes du nez , Qualité de vie , Rhinite , Sinusite , Humains , Anticorps monoclonaux humanisés/usage thérapeutique , Sinusite/traitement médicamenteux , Sinusite/complications , Polypes du nez/traitement médicamenteux , Polypes du nez/complications , Polypes du nez/immunologie , Asthme/traitement médicamenteux , Eczéma atopique/traitement médicamenteux , Eczéma atopique/complications , Rhinite/traitement médicamenteux , Oesophagite à éosinophiles/traitement médicamenteux , Maladie chronique , Mâle , Interleukine-13/antagonistes et inhibiteurs , Interleukine-13/immunologie , Multimorbidité , Interleukine-4/antagonistes et inhibiteurs , Interleukine-4/immunologie , Adulte , Femelle ,
20.
J Allergy Clin Immunol ; 154(2): 480-491, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38157943

RÉSUMÉ

BACKGROUND: The cytokine TSLP promotes type 2 immune responses and can induce adipose loss by stimulating lipid loss from the skin through sebum secretion by sebaceous glands, which enhances the skin barrier. However, the mechanism by which TSLP upregulates sebaceous gland function is unknown. OBJECTIVES: This study investigated the mechanism by which TSLP stimulates sebum secretion and adipose loss. METHODS: RNA-sequencing analysis was performed on sebaceous glands isolated by laser capture microdissection and single-cell RNA-sequencing analysis was performed on sorted skin T cells. Sebocyte function was analyzed by histological analysis and sebum secretion in vivo and by measuring lipogenesis and proliferation in vitro. RESULTS: This study found that TSLP sequentially stimulated the expression of lipogenesis genes followed by cell death genes in sebaceous glands to induce holocrine secretion of sebum. TSLP did not affect sebaceous gland activity directly. Rather, single-cell RNA-sequencing revealed that TSLP recruited distinct T-cell clusters that produce IL-4 and IL-13, which were necessary for TSLP-induced adipose loss and sebum secretion. Moreover, IL-13 was sufficient to cause sebum secretion and adipose loss in vivo and to induce lipogenesis and proliferation of a human sebocyte cell line in vitro. CONCLUSIONS: This study proposes that TSLP stimulates T cells to deliver IL-4 and IL-13 to sebaceous glands, which enhances sebaceous gland function, turnover, and subsequent adipose loss.


Sujet(s)
Cytokines , Interleukine-13 , Interleukine-4 , Glandes sébacées , Sébum , Lymphocytes T , Lymphopoïétine stromale thymique , Cytokines/métabolisme , Sébum/métabolisme , Sébum/immunologie , Interleukine-13/métabolisme , Interleukine-13/immunologie , Interleukine-4/métabolisme , Interleukine-4/immunologie , Animaux , Glandes sébacées/immunologie , Glandes sébacées/métabolisme , Lymphocytes T/immunologie , Humains , Souris , Tissu adipeux/immunologie , Tissu adipeux/métabolisme , Lipogenèse/immunologie , Souris de lignée C57BL
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE