Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Allergy Clin Immunol ; 152(5): 1121-1130.e10, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37277072

RESUMO

BACKGROUND: There is a need for new and effective oral asthma therapies. Dexpramipexole, an oral eosinophil-lowering drug, has not previously been studied in asthma. OBJECTIVE: We sought to evaluate the safety and efficacy of dexpramipexole in lowering blood and airway eosinophilia in subjects with eosinophilic asthma. METHODS: We performed a randomized, double-blind, placebo-controlled proof-of-concept trial in adults with inadequately controlled moderate to severe asthma and blood absolute eosinophil count (AEC) greater than or equal to 300/µL. Subjects were randomly assigned (1:1:1:1) to dexpramipexole 37.5, 75, or 150 mg BID (twice-daily) or placebo. The primary end point was the relative change in AEC from baseline to week 12. Prebronchodilator FEV1 week-12 change from baseline was a key secondary end point. Nasal eosinophil peroxidase was an exploratory end point. RESULTS: A total of 103 subjects were randomly assigned to dexpramipexole 37.5 mg BID (N = 22), 75 mg BID (N = 26), 150 mg BID (N = 28), or placebo (N = 27). Dexpramipexole significantly reduced placebo-corrected AEC week-12 ratio to baseline, in both the 150-mg BID (ratio, 0.23; 95% CI, 0.12-0.43; P < .0001) and the 75-mg BID (ratio, 0.34; 95% CI, 0.18-0.65; P = .0014) dose groups, corresponding to 77% and 66% reductions, respectively. Dexpramipexole reduced the exploratory end point of nasal eosinophil peroxidase week-12 ratio to baseline in the 150-mg BID (median, 0.11; P = .020) and the 75-mg BID (median, 0.17; P = .021) groups. Placebo-corrected FEV1 increases were observed starting at week 4 (nonsignificant). Dexpramipexole displayed a favorable safety profile. CONCLUSIONS: Dexpramipexole demonstrated effective eosinophil lowering and was well tolerated. Additional larger clinical trials are needed to understand the clinical efficacy of dexpramipexole in asthma.


Assuntos
Antiasmáticos , Asma , Eosinofilia Pulmonar , Adulto , Humanos , Pramipexol/farmacologia , Pramipexol/uso terapêutico , Peroxidase de Eosinófilo , Asma/tratamento farmacológico , Eosinofilia Pulmonar/tratamento farmacológico , Eosinófilos , Resultado do Tratamento , Método Duplo-Cego , Antiasmáticos/uso terapêutico
2.
Ann Allergy Asthma Immunol ; 130(1): 28-39, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36351516

RESUMO

Eosinophilic esophagitis (EoE) is a chronic and progressive immune-mediated disease of the esophagus associated with antigen-driven type 2 inflammation and symptoms of esophageal dysfunction. Our understanding of EoE pathophysiology has evolved since its initial recognition more than 20 years ago and has translated into diagnostic and novel therapeutic approaches that are affecting patient care. The mechanisms underlying disease development and progression are influenced by diverse factors, such as genetics, age, allergic comorbidities, and allergen exposures. Central to EoE pathophysiology is a dysregulated feed-forward cycle that develops between the esophageal epithelium and the immune system. Allergen-induced, type 2-biased immune activation by the esophageal epithelium propagates a cycle of impaired mucosal barrier integrity and allergic inflammation, eventually leading to tissue remodeling and progressive organ dysfunction. Herein, we review the current understanding of fundamental pathophysiological mechanisms contributing to EoE pathogenesis.


Assuntos
Esofagite Eosinofílica , Humanos , Esofagite Eosinofílica/diagnóstico , Inflamação , Eosinófilos
3.
J Allergy Clin Immunol ; 150(3): 666-675, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35413355

RESUMO

BACKGROUND: Virus-induced IFN-α secretion by plasmacytoid dendritic cells (pDCs) is negatively impacted by IgE and has been linked to asthma exacerbations. Eosinophils, another contributor to type 2 inflammation, are also associated with asthma severity. OBJECTIVE: We sought to investigate the impact of eosinophils on pDC antiviral interferon responses and determine whether anti-IL-5/5Rα therapy enhances pDC antiviral function. METHODS: Blood pDCs purified from anonymous donors were stimulated in vitro with rhinovirus (RV)-16 in the presence or absence of eosinophils/eosinophil supernatants. IFN-α was measured in supernatants and RNA collected for bulk RNA-sequencing. Next, purified pDCs from 8 individuals with moderate to severe asthma, treated or not treated with anti-IL-5/5Rα therapy, were cultured ex vivo with or without RV; IFN-α secretion and differential gene expression analysis were compared between groups. RESULTS: Exposure to either eosinophils or eosinophil supernatants inhibited RV-induced pDC IFN-α secretion in a dose-dependent manner and did not impact pDC viability. Eosinophil-derived neurotoxin and TGF-ß partially recapitulated pDC IFN-α inhibition. Transcriptome analysis revealed global repression of pDC interferon response patterns by eosinophils, most notably in basal expression of interferon-stimulated genes. Increased RV-induced IFN-α secretion and transcription as well as increased basal interferon-stimulated gene expression was detected in pDCs from participants treated with anti-IL-5/5Rα therapy. CONCLUSIONS: Our findings highlight a novel mechanism through which type 2 inflammation regulates pDC IFN-α responses relevant to RV respiratory infections in the context of eosinophilic airway disease, suggesting a potential mechanism through which eosinophil-depleting therapies may reduce severity of RV illnesses.


Assuntos
Asma , Eosinófilos , Antivirais/metabolismo , Asma/tratamento farmacológico , Asma/metabolismo , Células Dendríticas/metabolismo , Eosinófilos/metabolismo , Humanos , Inflamação/metabolismo , Interferon-alfa/metabolismo , Interleucina-5/imunologia , RNA/metabolismo , Rhinovirus/metabolismo
4.
J Allergy Clin Immunol ; 146(1): 1-7, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32344056

RESUMO

Eosinophils are circulating and tissue-resident leukocytes that have potent proinflammatory effects in a number of diseases. Recently, eosinophils have been shown to have various other functions, including immunoregulation and antiviral activity. Eosinophil levels vary dramatically in a number of clinical settings, especially following eosinophil-targeted therapy, which is now available to selectively deplete these cells. There are key coronavirus disease 2019 (COVID-19)-related questions concerning eosinophils whose answers affect recommended prevention and care. First, do patients with eosinophilia-associated diseases have an altered course of COVID-19? Second, do patients with eosinopenia (now intentionally induced by biological drugs) have unique COVID-19 susceptibility and/or disease course? This is a particularly relevant question because eosinopenia is associated with acute respiratory deterioration during infection with the severe acute respiratory syndrome coronavirus 2, the causative agent of COVID-19. Third, do eosinophils contribute to the lung pathology induced during COVID-19 and will they contribute to immunopotentiation potentially associated with emerging COVID-19 vaccines? Herein, we address these timely questions and project considerations during the emerging COVID-19 pandemic.


Assuntos
Betacoronavirus/imunologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Eosinófilos/imunologia , Pandemias/prevenção & controle , Pneumonia Viral/imunologia , Pneumonia Viral/prevenção & controle , Vacinas Virais/imunologia , Animais , COVID-19 , Vacinas contra COVID-19 , Humanos , SARS-CoV-2
5.
Curr Allergy Asthma Rep ; 20(8): 30, 2020 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-32506181

RESUMO

PURPOSE OF REVIEW: Recent research efforts have spurred great progress in the diagnosis and management of eosinophilic esophagitis (EoE). Nonetheless, challenges remain in addressing disease burden and impairment in the growing EoE population. We highlight work from the Cincinnati Center for Eosinophilic Disorders, the Consortium of Eosinophilic Gastrointestinal Disease Researchers, and others that address these ongoing challenges. RECENT FINDINGS: New tools for characterizing EoE disease activity include the EoE Histology Scoring System (EoEHSS), endoscopic alternatives, validated patient-reported outcome (PRO) questionnaires, and investigational biomarkers. These diagnostic and monitoring strategies have been complemented by advances in EoE therapy. Treatment modalities have refined the traditional approaches of dietary elimination, swallowed steroids, and proton pump inhibitors (PPI), and biologics offer promise for future treatment. This review summarizes EoE advances in disease management and newly defined EoE endotypes that may serve as the foundation for EoE-personalized medicine.


Assuntos
Esofagite Eosinofílica/terapia , Medicina de Precisão/métodos , Esofagite Eosinofílica/diagnóstico , Humanos
6.
J Pediatr Gastroenterol Nutr ; 70(4): 482-488, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31834110

RESUMO

OBJECTIVES: A minimally invasive biomarker to monitor disease activity is one of the greatest unmet clinical needs of the pediatric eosinophilic esophagitis (EoE) population. We aimed to determine whether circulating eosinophil progenitors (EoPs) could be used as a biomarker to identify pediatric patients with active EoE. METHODS: In a prospective observational study, peripheral blood samples, symptom history, and laboratory data were collected from pediatric patients undergoing endoscopy for evaluation of EoE on dietary therapy at Cincinnati Children's Hospital. Peripheral blood EoP level was determined by flow cytometry. RESULTS: Thirty-four children with active (n = 16) and inactive (n = 18) EoE were included in the analysis. EoP levels in the peripheral blood were 3-fold higher in patients with active EoE than inactive EoE (P < 0.0025). Blood absolute eosinophil count did not distinguish between active and inactive EoE (P = 0.16). A cut-off EoP level ≥17 accurately detected active disease in 79% of patients with 94.4% specificity and 62.5% sensitivity (area under the curve 0.81; P < 0.0024). Antihistamine use lowered the threshold EoP level to detect active EoE. CONCLUSIONS: This study suggests that blood EoP levels may be used as a biomarker to detect active EoE disease in patients undergoing food trials and potentially reduce the need for repeated endoscopies. Larger prospective studies are needed to investigate the effects of antihistamines and swallowed steroids on EoP mobilization into the peripheral blood and longitudinal studies to assess the performance of the assay in individual patients over time.


Assuntos
Esofagite Eosinofílica , Criança , Endoscopia , Esofagite Eosinofílica/diagnóstico , Eosinófilos , Humanos , Contagem de Leucócitos , Estudos Prospectivos
9.
J Immunol ; 189(6): 3064-77, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22888138

RESUMO

A fundamental step in the life cycle of Francisella tularensis is bacterial entry into host cells. F. tularensis activates complement, and recent data suggest that the classical pathway is required for complement factor C3 deposition on the bacterial surface. Nevertheless, C3 deposition is inefficient and neither the specific serum components necessary for classical pathway activation by F. tularensis in nonimmune human serum nor the receptors that mediate infection of neutrophils have been defined. In this study, human neutrophil uptake of GFP-expressing F. tularensis strains live vaccine strain and Schu S4 was quantified with high efficiency by flow cytometry. Using depleted sera and purified complement components, we demonstrated first that C1q and C3 were essential for F. tularensis phagocytosis, whereas C5 was not. Second, we used purification and immunodepletion approaches to identify a critical role for natural IgM in this process, and then used a wbtA2 mutant to identify LPS O-Ag and capsule as prominent targets of these Abs on the bacterial surface. Finally, we demonstrate using receptor-blocking Abs that CR1 (CD35) and CR3 (CD11b/CD18) acted in concert for phagocytosis of opsonized F. tularensis by human neutrophils, whereas CR3 and CR4 (CD11c/CD18) mediated infection of human monocyte-derived macrophages. Altogether, our data provide fundamental insight into mechanisms of F. tularensis phagocytosis and support a model whereby natural IgM binds to surface capsular and O-Ag polysaccharides of F. tularensis and initiates the classical complement cascade via C1q to promote C3 opsonization of the bacterium and phagocytosis via CR3 and either CR1 or CR4 in a phagocyte-specific manner.


Assuntos
Francisella tularensis/imunologia , Soros Imunes/fisiologia , Imunoglobulina M/fisiologia , Antígeno de Macrófago 1/fisiologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Receptores de Complemento 3b/fisiologia , Receptores de Complemento/fisiologia , Adulto , Animais , Francisella tularensis/metabolismo , Humanos , Imunoglobulina M/sangue , Neutrófilos/metabolismo , Proteínas Opsonizantes/metabolismo , Fagocitose/imunologia , Ovinos
10.
J Immunol ; 188(7): 3351-63, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22357630

RESUMO

Francisella tularensis is a facultative intracellular bacterium that infects many cell types, including neutrophils. We demonstrated previously that F. tularensis inhibits NADPH oxidase assembly and activity and then escapes the phagosome to the cytosol, but effects on other aspects of neutrophil function are unknown. Neutrophils are short-lived cells that undergo constitutive apoptosis, and phagocytosis typically accelerates this process. We now demonstrate that F. tularensis significantly inhibited neutrophil apoptosis as indicated by morphologic analysis as well as annexin V and TUNEL staining. Thus, ∼80% of infected neutrophils remained viable at 48 h compared with ∼50% of control cells, and ∼40% of neutrophils that ingested opsonized zymosan. In keeping with this finding, processing and activation of procaspases-8, -9, and -3 were markedly diminished and delayed. F. tularensis also significantly impaired apoptosis triggered by Fas crosslinking. Of note, these effects were dose dependent and could be conferred by either intracellular or extracellular live bacteria, but not by formalin-killed organisms or isolated LPS and capsule, and were not affected by disruption of wbtA2 or FTT1236/FTL0708-genes required for LPS O-antigen and capsule biosynthesis. In summary, we demonstrate that F. tularensis profoundly impairs constitutive neutrophil apoptosis via effects on the intrinsic and extrinsic pathways, and thereby define a new aspect of innate immune evasion by this organism. As defects in neutrophil turnover prevent resolution of inflammation, our findings also suggest a mechanism that may in part account for the neutrophil accumulation, granuloma formation, and severe tissue damage that characterizes lethal pneumonic tularemia.


Assuntos
Apoptose/fisiologia , Francisella tularensis/fisiologia , Evasão da Resposta Imune/imunologia , Neutrófilos/microbiologia , Adulto , Anexina A5/análise , Cápsulas Bacterianas/genética , Cápsulas Bacterianas/imunologia , Caspases/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Fragmentação do DNA , Ativação Enzimática , Francisella tularensis/genética , Francisella tularensis/imunologia , Francisella tularensis/patogenicidade , Humanos , Marcação In Situ das Extremidades Cortadas , Interleucina-8/análise , Lipopolissacarídeos/imunologia , Neutrófilos/imunologia , Neutrófilos/patologia , Proteínas Opsonizantes/imunologia , Fagocitose , Explosão Respiratória , Virulência , Zimosan/imunologia , Receptor fas/fisiologia
11.
J Allergy Clin Immunol Pract ; 12(5): 1159-1169, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38423284

RESUMO

Oral immunotherapy (OIT) is an accessible procedure for practicing allergy/immunology providers, yet rigorous safety standards are limited in the clinical setting. By exploring the transition from research to clinical practice OIT, we review relevant safety considerations necessary for the clinical provider. We offer a perspective on clinical benefits and considerations at the individual, collaboration, and policy levels from the vantage of a large academic OIT program, and we propose several practical start-up checklists and clerical considerations for practicing providers. Awareness of the local population and front-end planning is necessary to improve the accessibility of this procedure in clinical practice among racial and socioeconomic minority populations. Sharing and merging OIT protocols, procedural methods, and electronic medical record order sets may increase harmonization among OIT-providing institutions and further our abilities to pool safety and outcomes data, ultimately enhancing the safety and efficacy of clinical OIT.


Assuntos
Dessensibilização Imunológica , Hipersensibilidade Alimentar , Humanos , Dessensibilização Imunológica/métodos , Hipersensibilidade Alimentar/terapia , Administração Oral , Estados Unidos , Centros Médicos Acadêmicos , Alérgenos/imunologia , Alérgenos/administração & dosagem
12.
Artigo em Inglês | MEDLINE | ID: mdl-38851488

RESUMO

The OIT-BRAVE questionnaire was developed to serve as a clinical screening tool to identify patients who may be experiencing adverse effects with oral immunotherapy.

13.
J Allergy Clin Immunol Pract ; 11(5): 1509-1521.e6, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36801492

RESUMO

BACKGROUND: Eosinophilic esophagitis (EoE) can coexist in individuals with food allergy. OBJECTIVE: To evaluate the characteristics of food-allergic patients with and without coexisting EoE using a large food allergy patient registry. METHODS: Data were derived from 2 Food Allergy Research & Education, Inc, Patient Registry surveys. A series of multivariable regression models were used to evaluate associations between demographic, comorbidity, and food allergy characteristics and the likelihood of reporting EoE. RESULTS: Five percent (n = 309) of registry participants (n = 6074; ages <1 year->80 years, mean, 20.20 ± 15.37 years) reported having EoE. The odds of having EoE were significantly greater in male participants (adjusted odds ratio [aOR], 1.3; 95% CI, 1.04-1.72) and those with comorbid asthma (aOR, 2.0; 95% CI, 1.55-2.49), allergic rhinitis (aOR, 1.8; 95% CI, 1.37-2.22), oral allergy syndrome (aOR, 2.8; 95% CI, 2.09-3.70), food protein-induced enterocolitis syndrome (aOR, 2.5; 95% CI, 1.34-4.84), and hyper-IgE syndrome (aOR, 7.6; 95% CI, 2.93-19.92), though not atopic dermatitis (aOR, 1.3; 95% CI, 0.99-1.59), when adjusting for demographics (sex, age, race, ethnicity, and geographic location). Those with a greater number of food allergies (aOR, 1.3; 95% CI, 1.23-1.32), more frequent food-related allergic reactions (aOR, 1.2; 95% CI, 1.11-1.24), previous anaphylaxis (aOR, 1.5; 95% CI, 1.15-1.83), and health care utilization for food-related allergic reactions (aOR, 1.3; 95% CI, 1.01-1.67)-specifically intensive care unit admission (aOR, 1.2; 95% CI, 1.07-1.33)-were more likely to have EoE after controlling for demographics. However, no significant difference in ever using epinephrine for food-related allergic reactions was detected. CONCLUSIONS: These self-reported data showed that coexisting EoE is associated with an increased number of food allergies, food-related allergic reactions per year, and measures of reaction severity, calling attention to the likely increased health care needs of food-allergic patients with EoE.


Assuntos
Asma , Esofagite Eosinofílica , Hipersensibilidade Alimentar , Rinite Alérgica , Humanos , Masculino , Lactente , Feminino , Esofagite Eosinofílica/epidemiologia , Hipersensibilidade Alimentar/epidemiologia , Hipersensibilidade Alimentar/complicações , Asma/complicações , Alérgenos , Sistema de Registros
14.
bioRxiv ; 2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36778284

RESUMO

The skin is a major immune organ and skin barrier dysfunction is a major risk factor for the development of the inappropriate immune response seen in allergic disease. Skin barrier disruption alters the landscape of antigens experienced by the immune system and the downstream impacts on the antibody repertoire remain poorly characterized, particularly for the IgE isotype responsible for allergic specificity and in early life, when allergic disease is developing. In this study, we sequenced antibody gene repertoires from a large and well-characterized cohort of children with atopic dermatitis and found that food sensitization was associated with lower mutation frequencies in the IgE compartment. This trend was abrogated in children living with pets during the first year of life. These results elucidate potential molecular mechanisms underlying the protective effects of pet ownership and non-antiseptic environs reported for allergic disease, and the hygiene hypothesis more broadly. We also observed increased IgE diversity and increased isotype-switching to the IgE isotype, suggesting that B cell development, particularly isotype-switching, is heavily altered in the those with food allergen sensitizations relative to those without food allergen sensitizations. Unlike for food antigens, aeroallergen sensitization exhibited no effect on IgE mutation or diversity. Consistent patterns of antibody rearrangement were associated with food allergen sensitization in subjects with atopic dermatitis. Thus, we propose the Immune Repertoire in Atopic Disease (IRAD) score, to quantify this repertoire shift and to aid clinically in patient diagnosis and risk stratification.

15.
Lancet Gastroenterol Hepatol ; 8(9): 803-815, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37336228

RESUMO

BACKGROUND: In eosinophilic gastrointestinal diseases, the role of eosinophils in disease pathogenesis and the effect of eosinophil depletion on patient outcomes are unclear. Benralizumab, an eosinophil-depleting monoclonal antibody that targets the interleukin-5 receptor α, might eliminate gastric tissue eosinophils and improve outcomes in eosinophilic gastritis. We aimed to assess the efficacy and safety of benralizumab in patients with eosinophilic gastritis. METHODS: We conducted a single-site, randomised, double-blind, placebo-controlled, phase 2 trial at Cincinnati Children's Hospital Medical Center (Cincinnati, OH, USA). Individuals aged 12-60 years with symptomatic, histologically active eosinophilic gastritis (peak gastric eosinophil count ≥30 eosinophils per high-power field [eos/hpf] in at least five hpfs) and blood eosinophilia (>500 eosinophils per µL [eos/µL]) were randomly assigned (1:1, block size of four) to benralizumab 30 mg or placebo, stratified by the use of glucocorticoids for gastric disease. Investigators, study staff, and study participants were masked to treatment assignment; statisticians were unmasked when analysing data. Treatments were administered subcutaneously once every 4 weeks for a 12-week double-blind period (three total injections). The primary endpoint was the proportion of patients who achieved histological remission (peak gastric eosinophil count <30 eos/hpf) at week 12. Key secondary endpoints were the changes from baseline to week 12 in peak gastric eosinophil count, blood eosinophil count, eosinophilic gastritis histology (total, inflammatory, and structural feature scores), Eosinophilic Gastritis Endoscopic Reference System (EG-REFS) score, and patient-reported outcome symptom measures (Severity of Dyspepsia Assessment [SODA] and Patient-Reported Outcome Measurement Information System [PROMIS] short-form questionnaire). After the 12-week double-blind period, patients were eligible for entry into two open-label extension (OLE) periods up to week 88, in which all patients received benralizumab. Efficacy was analysed in the intention-to-treat (ITT) population and safety was assessed in all patients who received at least one dose of study drug. The trial was registered on ClinicalTrials.gov, NCT03473977, and is completed. FINDINGS: Between April 23, 2018, and Jan 13, 2020, 34 patients were screened, and 26 were subsequently randomly assigned to benralizumab (n=13) or placebo (n=13) and included in the ITT and safety populations (mean age 19·5 years [SD 7·3]; 19 [73%] male patients and seven [27%] female patients). At week 12, ten (77% [95% CI 50 to 92]) of 13 patients who received benralizumab and one (8% [1 to 33]) of 13 who received placebo achieved histological remission (difference 69 percentage points [95% CI 32 to 85]; p=0·0010). Changes from baseline to week 12 were significantly greater in the benralizumab group versus the placebo group for peak gastric eosinophil counts (mean -137 eos/hpf [95% CI -186 to -88] vs -38 eos/hpf [-94 to 18]; p=0·0080), eosinophilic gastritis histology total score (mean -0·31 [-0·42 to -0·20] vs -0·02 [-0·16 to 0·12]; p=0·0016), histology inflammatory score (mean -0·46 [-0·60 to -0·31] vs -0·04 [-0·22 to 0·13]; p=0·0006), and blood eosinophil counts (median -1060 eos/µL [IQR -1740 to -830] vs -160 eos/µL [-710 to 120]; p=0·0044). Changes were not significantly different between the groups for eosinophilic gastritis histology structural score (mean -0·07 [95% CI -0·19 to 0·05] vs 0·03 [-0·09 to 0·15]; p=0·23), EG-REFS score (mean -1·0 [-2·3 to 0·3] vs -0·5 [-2·0 to 1·0]; p=0·62), or in patient-reported outcomes (SODA and PROMIS). During the double-blind period, treatment-emergent adverse events occurred in 11 (85%) of 13 patients in the benralizumab group and six (46%) of 13 in the placebo group; the most common treatment-emergent adverse events were headache (six [46%] vs two [15%] patients), nausea (three [23%] vs two [15%]), and vomiting (two [15%] vs three [23%]). There were no treatment-related deaths. Two patients had serious adverse events (dizziness and rhabdomyolysis in one patient; aspiration in one patient) during the OLE periods, which were considered unrelated to study treatment. INTERPRETATION: Benralizumab treatment induced histological remission, as defined by absence of tissue eosinophilia, in most patients with eosinophilic gastritis. However, the persistence of histological, endoscopic, and other features of the disease suggest a co-existing, eosinophil-independent pathogenic mechanism and the need for broader targeting of type 2 immunity. FUNDING: AstraZeneca and the Division of Intramural Research (National Institute of Allergy and Infectious Diseases, US National Institutes of Health).


Assuntos
Asma , Eosinofilia , Estados Unidos , Criança , Humanos , Masculino , Feminino , Adulto Jovem , Adulto , Asma/complicações , Asma/tratamento farmacológico , Progressão da Doença , Eosinofilia/tratamento farmacológico
16.
Mucosal Immunol ; 14(6): 1271-1281, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34341502

RESUMO

Expression of Ikaros family transcription factor IKZF3 (Aiolos) increases during murine eosinophil lineage commitment and maturation. Herein, we investigated Aiolos expression and function in mature human and murine eosinophils. Murine eosinophils deficient in Aiolos demonstrated gene expression changes in pathways associated with granulocyte-mediated immunity, chemotaxis, degranulation, ERK/MAPK signaling, and extracellular matrix organization; these genes had ATAC peaks within 1 kB of the TSS that were enriched for Aiolos-binding motifs. Global Aiolos deficiency reduced eosinophil frequency within peripheral tissues during homeostasis; a chimeric mouse model demonstrated dependence on intrinsic Aiolos expression by eosinophils. Aiolos deficiency reduced eosinophil CCR3 surface expression, intracellular ERK1/2 signaling, and CCL11-induced actin polymerization, emphasizing an impaired functional response. Aiolos-deficient eosinophils had reduced tissue accumulation in chemokine-, antigen-, and IL-13-driven inflammatory experimental models, all of which at least partially depend on CCR3 signaling. Human Aiolos expression was associated with active chromatin marks enriched for IKZF3, PU.1, and GATA-1-binding motifs within eosinophil-specific histone ChIP-seq peaks. Furthermore, treating the EOL-1 human eosinophilic cell line with lenalidomide yielded a dose-dependent decrease in Aiolos. These collective data indicate that eosinophil homing during homeostatic and inflammatory allergic states is Aiolos-dependent, identifying Aiolos as a potential therapeutic target for eosinophilic disease.


Assuntos
Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Eosinófilos/imunologia , Eosinófilos/metabolismo , Fator de Transcrição Ikaros/genética , Alérgenos/imunologia , Animais , Suscetibilidade a Doenças , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Granulócitos/imunologia , Granulócitos/metabolismo , Humanos , Fator de Transcrição Ikaros/metabolismo , Imunidade Inata , Imunofenotipagem , Contagem de Leucócitos , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Receptores CCR3/genética , Receptores CCR3/metabolismo , Transdução de Sinais
17.
J Allergy Clin Immunol Pract ; 7(8): 2750-2758.e2, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31128377

RESUMO

BACKGROUND: Eosinophilia is associated with various conditions, including allergic, infectious, and neoplastic disorders. The diagnostic differential is broad, and data on hypereosinophilia in pediatric patients are limited. OBJECTIVE: The objectives of this study were to identify cases of hypereosinophilia in a tertiary pediatric medical center, determine clinical characteristics and disease associations, and estimate the incidence of hypereosinophilia in the hospital and geographic populations. METHODS: A retrospective chart review included patients younger than 18 years presenting to a tertiary pediatric medical center (January 1, 2008, to May 31, 2017) with absolute eosinophil counts (AECs) greater than or equal to 1.50 thousand eosinophils/microliter (K/µL) recorded on at least 2 occasions at least 4 weeks apart (N = 176). Clinical characteristics, laboratory values, treatment course, and associated diagnoses were evaluated. RESULTS: The most common cause of hypereosinophilia in this cohort was secondary hypereosinophilia. Atopic dermatitis, graft-versus-host disease, sickle cell disease, and parasitic infections were the most common conditions associated with hypereosinophilia. Median age at diagnosis was 4.6 (interquartile range, 1.5-10.5) years. Median peak AEC was 3.16 (2.46-4.78) K/µL. Hypereosinophilia occurred most frequently in patients aged between 6 and 11 years (24.4%) and younger than 1 year (18.2%). Patients with neoplasms and immune deficiencies had significantly higher peak AECs than did patients with overlap hypereosinophilic syndrome and atopic diseases (P < .0001). CONCLUSIONS: Pediatric hypereosinophilia has an incidence of 54.4 per 100,000 persons per year, with children younger than 1 year and aged 6 to 11 years accounting for most affected patients. Pediatric hypereosinophilia is not uncommon and remains underrecognized, highlighting a need for clinicians to identify patients who meet criteria for hypereosinophilia and to pursue a thorough evaluation.


Assuntos
Eosinofilia/epidemiologia , Anemia Falciforme/complicações , Anemia Falciforme/epidemiologia , Criança , Pré-Escolar , Dermatite Atópica/complicações , Dermatite Atópica/epidemiologia , Eosinofilia/etiologia , Feminino , Doença Enxerto-Hospedeiro/complicações , Doença Enxerto-Hospedeiro/epidemiologia , Hospitais Pediátricos , Humanos , Incidência , Lactente , Masculino , Doenças Parasitárias/complicações , Doenças Parasitárias/epidemiologia , Estudos Retrospectivos , Centros de Atenção Terciária
18.
Front Immunol ; 9: 1944, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30233571

RESUMO

Hypereosinophilia (HE) is currently defined by a peripheral blood absolute eosinophil count (AEC) of ≥1,500 cells/microL. Although mild blood eosinophilia (AEC 500-1,500 cells/microL) is observed relatively frequently within the pediatric population, persistent HE is uncommon and should prompt additional clinical evaluation. While the clinical manifestations and underlying etiologies of HE in adults have been well-characterized, there is a paucity of data on HE in children. Limited evidence suggests that many similarities between adult and pediatric HE likely exist, but some important differences remain between these populations. The evaluation of HE in children can be challenging given the broad differential diagnosis, which includes primary hematologic disorders and secondary eosinophilia in which the increased eosinophil levels are propagated by disease states that promote eosinophil production and survival. On the basis of the underlying etiology, clinical manifestations can range from benign, self-resolving elevations in the AEC to life-threatening disorders with the potential for significant end-organ damage. Given the broad differential diagnosis of HE, it remains essential to systematically approach the evaluation of unexplained HE in children. This review will discuss the differential diagnosis for pediatric HE, highlighting etiologies that are more prevalent within the pediatric population. Additionally, a summary of the epidemiology of pediatric HE will be presented, with focus on some of the differences that exist between pediatric and adult HE. Finally, a directed approach to the diagnostic evaluation of children with HE will be discussed.


Assuntos
Síndrome Hipereosinofílica/diagnóstico , Síndrome Hipereosinofílica/imunologia , Adolescente , Adulto , Criança , Pré-Escolar , Diagnóstico Diferencial , Feminino , Humanos , Síndrome Hipereosinofílica/sangue , Síndrome Hipereosinofílica/terapia , Lactente , Recém-Nascido , Contagem de Leucócitos , Masculino
19.
J Leukoc Biol ; 79(6): 1214-25, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16543403

RESUMO

Previous studies have demonstrated that Helicobacter pylori (Hp) delays its entry into macrophages and persists inside megasomes, which are poorly acidified and accumulate early endosome autoantigen 1. Herein, we explored the role of Hp urease in bacterial survival in murine peritoneal macrophages and J774 cells. Plasmid-free mutagenesis was used to replace ureA and ureB with chloramphenicol acetyltransferase in Hp Strains 11637 and 11916. ureAB null Hp lacked detectable urease activity and did not express UreA or UreB as judged by immunoblotting. Deletion of ureAB had no effect on Hp binding to macrophages or the rate or extent of phagocytosis. However, intracellular survival of mutant organisms was impaired significantly. Immunofluorescence microscopy demonstrated that (in contrast to parental organisms) mutant Hp resided in single phagosomes, which were acidic and accumulated the lysosome marker lysosome-associated membrane protein-1 but not early endosome autoantigen 1. A similar phenotype was observed for spontaneous urease mutants derived from Hp Strain 60190. Treatment of macrophages with bafilomycin A1, NH4Cl, or chloroquine prevented acidification of phagosomes containing mutant Hp. However, only ammonium chloride enhanced bacterial viability significantly. Rescue of ureAB null organisms was also achieved by surface adsorption of active urease. Altogether, our data indicate a role for urease and urease-derived ammonia in megasome formation and Hp survival.


Assuntos
Helicobacter pylori/enzimologia , Macrófagos/microbiologia , Fagossomos/fisiologia , Urease/fisiologia , Adsorção , Amônia/metabolismo , Cloreto de Amônio/farmacologia , Animais , Autoantígenos/análise , Linhagem Celular/microbiologia , Cloroquina/farmacologia , Indução Enzimática , Feminino , Deleção de Genes , Genes Reporter , Helicobacter pylori/classificação , Helicobacter pylori/genética , Helicobacter pylori/fisiologia , Concentração de Íons de Hidrogênio , Proteínas de Membrana Lisossomal/análise , Macrolídeos/farmacologia , Macrófagos Peritoneais/microbiologia , Fusão de Membrana , Proteínas de Membrana/análise , Camundongos , Microscopia de Fluorescência , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Fagocitose , Fagossomos/química , Fagossomos/microbiologia , Fenótipo , Especificidade da Espécie , Urease/genética , Urease/farmacologia , Proteínas de Transporte Vesicular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA