Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 235
Filter
1.
J Clin Oncol ; 41(36): 5482-5492, 2023 12 20.
Article in English | MEDLINE | ID: mdl-38100992

ABSTRACT

PURPOSE: Most patients with advanced pancreas cancer experience pain and must limit their daily activities because of tumor-related symptoms. To date, no treatment has had a significant impact on the disease. In early studies with gemcitabine, patients with pancreas cancer experienced an improvement in disease-related symptoms. Based on those findings, a definitive trial was performed to assess the effectiveness of gemcitabine in patients with newly diagnosed advanced pancreas cancer. PATIENTS AND METHODS: One hundred twenty-six patients with advanced symptomatic pancreas cancer completed a lead-in period to characterize and stabilize pain and were randomized to receive either gemcitabine 1,000 mg/m2 weekly x 7 followed by 1 week of rest, then weekly x 3 every 4 weeks thereafter (63 patients), or to fluorouracil (5-FU) 600 mg/m2 once weekly (63 patients). The primary efficacy measure was clinical benefit response, which was a composite of measurements of pain (analgesic consumption and pain intensity), Karnofsky performance status, and weight. Clinical benefit required a sustained (> or = 4 weeks) improvement in at least one parameter without worsening in any others. Other measures of efficacy included response rate, time to progressive disease, and survival. RESULTS: Clinical benefit response was experienced by 23.8% of gemcitabine-treated patients compared with 4.8% of 5-FU-treated patients (P = .0022). The median survival durations were 5.65 and 4.41 months for gemcitabine-treated and 5-FU-treated patients, respectively (P = .0025). The survival rate at 12 months was 18% for gemcitabine patients and 2% for 5-FU patients. Treatment was well tolerated. CONCLUSION: This study demonstrates that gemcitabine is more effective than 5-FU in alleviation of some disease-related symptoms in patients with advanced, symptomatic pancreas cancer. Gemcitabine also confers a modest survival advantage over treatment with 5-FU.

3.
Allergy ; 78(5): 1169-1203, 2023 05.
Article in English | MEDLINE | ID: mdl-36799120

ABSTRACT

Asthma, rhinitis, and atopic dermatitis (AD) are interrelated clinical phenotypes that partly overlap in the human interactome. The concept of "one-airway-one-disease," coined over 20 years ago, is a simplistic approach of the links between upper- and lower-airway allergic diseases. With new data, it is time to reassess the concept. This article reviews (i) the clinical observations that led to Allergic Rhinitis and its Impact on Asthma (ARIA), (ii) new insights into polysensitization and multimorbidity, (iii) advances in mHealth for novel phenotype definitions, (iv) confirmation in canonical epidemiologic studies, (v) genomic findings, (vi) treatment approaches, and (vii) novel concepts on the onset of rhinitis and multimorbidity. One recent concept, bringing together upper- and lower-airway allergic diseases with skin, gut, and neuropsychiatric multimorbidities, is the "Epithelial Barrier Hypothesis." This review determined that the "one-airway-one-disease" concept does not always hold true and that several phenotypes of disease can be defined. These phenotypes include an extreme "allergic" (asthma) phenotype combining asthma, rhinitis, and conjunctivitis. Rhinitis alone and rhinitis and asthma multimorbidity represent two distinct diseases with the following differences: (i) genomic and transcriptomic background (Toll-Like Receptors and IL-17 for rhinitis alone as a local disease; IL-33 and IL-5 for allergic and non-allergic multimorbidity as a systemic disease), (ii) allergen sensitization patterns (mono- or pauci-sensitization versus polysensitization), (iii) severity of symptoms, and (iv) treatment response. In conclusion, rhinitis alone (local disease) and rhinitis with asthma multimorbidity (systemic disease) should be considered as two distinct diseases, possibly modulated by the microbiome, and may be a model for understanding the epidemics of chronic and autoimmune diseases.


Subject(s)
Asthma , Rhinitis, Allergic , Rhinitis , Humans , Rhinitis/diagnosis , Rhinitis/epidemiology , Rhinitis/complications , Asthma/diagnosis , Asthma/epidemiology , Asthma/etiology , Rhinitis, Allergic/complications , Allergens , Multimorbidity
4.
Allergy ; 73(9): 1892-1901, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29790577

ABSTRACT

BACKGROUND: Recent data associate eosinophilic esophagitis (EoE) with IgG4 rather than IgE, but its significance and function have not been determined. Our aims were to measure esophageal IgG4 levels and to determine functional correlations as assessed by histologic and transcriptome analyses. METHODS: This case-control study included pediatric subjects with EoE (≥15 eosinophils/HPF) and non-EoE controls. Protein lysates were analyzed for IgA, IgM, and IgG1-IgG4 using the Luminex 100 system; IgE was quantified by ELISA. Esophageal biopsies were scored using the EoE histology scoring system. Transcripts were probed by the EoE diagnostic panel, designed to examine the expression of 96 esophageal transcripts. RESULTS: Esophageal IgG subclasses, IgA, and IgM, but not IgE, were increased in subjects with EoE relative to controls. The greatest change between groups was seen in IgG4 (4.2 mg/g protein [interquartile range: 1.0-13.1 mg/g protein] vs 0.2 mg/g protein [0.1-0.9]; P < .0001). Tissue IgG4 levels correlated with esophageal eosinophil counts (P = .0006); histologic grade (P = .0011) and stage (P = .0112) scores; and IL4, IL10, IL13, but not TGFB1, expression and had strong associations with a subset of the EoE transcriptome. Esophageal IgG4 transcript expression was increased and correlated with IgG4 protein levels and IL10 expression. CONCLUSION: These findings extend prior studies on IgG4 in adult EoE to the pediatric population and provide deeper understanding of the potential significance and regulation of IgG4, demonstrating that IgG4 is a relevant feature of the disease; is closely related to esophageal eosinophil levels, type 2 immunity and T regulatory cytokines; and is likely produced locally.


Subject(s)
Eosinophilic Esophagitis/diagnosis , Eosinophilic Esophagitis/etiology , Immunoglobulin G/immunology , Transcriptome , Biopsy , Case-Control Studies , Child , Child, Preschool , Esophageal Mucosa/immunology , Esophageal Mucosa/metabolism , Esophageal Mucosa/pathology , Esophagus/immunology , Esophagus/metabolism , Esophagus/pathology , Female , Gene Expression , Histocytochemistry , Humans , Immunoglobulin G/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Isotypes/immunology , Male
5.
Sci Rep ; 7(1): 17563, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29242581

ABSTRACT

The molecular and cellular etiology of eosinophilic esophagitis (EoE), an emerging tissue-specific allergic disease, involves dysregulated gene expression in esophageal epithelial cells. Herein, we assessed the esophageal expression of IL-33, an epithelium-derived alarmin cytokine, in patients with EoE. IL-33 protein was markedly overexpressed within the nuclei of a subpopulation of basal layer esophageal epithelial cells in patients with active EoE compared to control individuals. IL-33 exhibited dynamic expression as levels normalized upon EoE remission. IL-33-positive basal epithelial cells expressed E-cadherin and the undifferentiated epithelial cell markers keratin 5 and 14 but not the differentiation marker keratin 4. Moreover, the IL-33-positive epithelial cells expressed the epithelial progenitor markers p75 and p63 and lacked the proliferation markers Ki67 and phospho-histone H3. Additionally, the IL-33-positive cells had low expression of PCNA. IL-33 expression was detected in ex vivo-cultured primary esophageal epithelial cells in a subpopulation of cells lacking expression of proliferation markers. Collectively, we report that IL-33 expression is induced in an undifferentiated, non-dividing esophageal epithelial cell population in patients with active EoE.


Subject(s)
Eosinophilic Esophagitis/metabolism , Eosinophilic Esophagitis/pathology , Epithelial Cells/metabolism , Esophagus/pathology , Interleukin-33/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Humans
8.
Mucosal Immunol ; 10(3): 580-588, 2017 05.
Article in English | MEDLINE | ID: mdl-28224995

ABSTRACT

Eosinophilic esophagitis (EoE) is a chronic, allergic disease associated with marked mucosal eosinophil accumulation. EoE disease risk is multifactorial and includes environmental and genetic factors. This review will focus on the contribution of genetic variation to EoE risk, as well as the experimental tools and statistical methodology used to identify EoE risk loci. Specific disease-risk loci that are shared between EoE and other allergic diseases (TSLP, LRRC32) or unique to EoE (CAPN14), as well as Mendellian Disorders associated with EoE, will be reviewed in the context of the insight that they provide into the molecular pathoetiology of EoE. We will also discuss the clinical opportunities that genetic analyses provide in the form of decision support tools, molecular diagnostics, and novel therapeutic approaches.


Subject(s)
Calpain/genetics , Cytokines/genetics , Eosinophilic Esophagitis/genetics , Membrane Proteins/genetics , Mucous Membrane/immunology , Animals , Clinical Decision-Making , Eosinophils , Gene-Environment Interaction , Genetic Predisposition to Disease , Genetic Testing , Genotype , Humans , Polymorphism, Genetic , Thymic Stromal Lymphopoietin
9.
J Exp Med ; 214(3): 669-680, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28126831

ABSTRACT

Nonimmunological connective tissue phenotypes in humans are common among some congenital and acquired allergic diseases. Several of these congenital disorders have been associated with either increased TGF-ß activity or impaired STAT3 activation, suggesting that these pathways might intersect and that their disruption may contribute to atopy. In this study, we show that STAT3 negatively regulates TGF-ß signaling via ERBB2-interacting protein (ERBIN), a SMAD anchor for receptor activation and SMAD2/3 binding protein. Individuals with dominant-negative STAT3 mutations (STAT3mut ) or a loss-of-function mutation in ERBB2IP (ERBB2IPmut ) have evidence of deregulated TGF-ß signaling with increased regulatory T cells and total FOXP3 expression. These naturally occurring mutations, recapitulated in vitro, impair STAT3-ERBIN-SMAD2/3 complex formation and fail to constrain nuclear pSMAD2/3 in response to TGF-ß. In turn, cell-intrinsic deregulation of TGF-ß signaling is associated with increased functional IL-4Rα expression on naive lymphocytes and can induce expression and activation of the IL-4/IL-4Rα/GATA3 axis in vitro. These findings link increased TGF-ß pathway activation in ERBB2IPmut and STAT3mut patient lymphocytes with increased T helper type 2 cytokine expression and elevated IgE.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Hypersensitivity/immunology , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Adaptor Proteins, Signal Transducing/deficiency , Humans , Interleukin-4/physiology , Receptors, Interleukin-4/physiology , Smad2 Protein/analysis , Smad2 Protein/physiology , Smad3 Protein/analysis , Smad3 Protein/physiology
10.
Mucosal Immunol ; 10(5): 1190-1201, 2017 09.
Article in English | MEDLINE | ID: mdl-28051089

ABSTRACT

Cadherins (CDH) mediate diverse processes critical in inflammation, including cell adhesion, migration, and differentiation. Herein, we report that the uncharacterized cadherin 26 (CDH26) is highly expressed by epithelial cells in human allergic gastrointestinal tissue. In vitro, CDH26 promotes calcium-dependent cellular adhesion of cells lacking endogenous CDHs by a mechanism involving homotypic binding and interaction with catenin family members (alpha, beta, and p120), as assessed by biochemical assays. Additionally, CDH26 enhances cellular adhesion to recombinant integrin α4ß7 in vitro; conversely, recombinant CDH26 binds αE and α4 integrins in biochemical and cellular functional assays, respectively. Interestingly, CDH26-Fc inhibits activation of human CD4+ T cells in vitro including secretion of IL-2. Taken together, we have identified a novel functional CDH regulated during allergic responses with unique immunomodulatory properties, as it binds α4 and αE integrins and regulates leukocyte adhesion and activation, and may thus represent a novel checkpoint for immune regulation and therapy via CDH26-Fc.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cadherins/metabolism , Epithelial Cells/physiology , Hypersensitivity/immunology , Inflammation/immunology , Intestinal Mucosa/metabolism , Adolescent , Adult , Cadherins/genetics , Cell Adhesion , Child , Child, Preschool , Female , HEK293 Cells , Humans , Infant , Integrin alpha Chains/metabolism , Integrin alpha4/metabolism , Integrin beta Chains/metabolism , Intestines/pathology , Lymphocyte Activation , Male , Protein Binding , Young Adult
11.
Dis Esophagus ; 30(3): 1-8, 2017 02 01.
Article in English | MEDLINE | ID: mdl-26857345

ABSTRACT

Eosinophilic esophagitis (EoE) is diagnosed by symptoms, and at least 15 intraepithelial eosinophils per high power field in an esophageal biopsy. Other pathologic features have not been emphasized. We developed a histology scoring system for esophageal biopsies that evaluates eight features: eosinophil density, basal zone hyperplasia, eosinophil abscesses, eosinophil surface layering, dilated intercellular spaces (DIS), surface epithelial alteration, dyskeratotic epithelial cells, and lamina propria fibrosis. Severity (grade) and extent (stage) of abnormalities were scored using a 4-point scale (0 normal; 3 maximum change). Reliability was demonstrated by strong to moderate agreement among three pathologists who scored biopsies independently (P ≤ 0.008). Several features were often abnormal in 201 biopsies (101 distal, 100 proximal) from 104 subjects (34 untreated, 167 treated). Median grade and stage scores were significantly higher in untreated compared with treated subjects (P ≤ 0.0062). Grade scores for features independent of eosinophil counts were significantly higher in biopsies from untreated compared with treated subjects (basal zone hyperplasia P ≤ 0.024 and DIS P ≤ 0.005), and were strongly correlated (R-square >0.67). Principal components analysis identified three principal components that explained 78.2% of the variation in the features. In logistic regression models, two principal components more closely associated with treatment status than log distal peak eosinophil count (PEC) (R-square 17, area under the curve (AUC) 77.8 vs. R-square 9, AUC 69.8). In summary, the EoE histology scoring system provides a method to objectively assess histologic changes in the esophagus beyond eosinophil number. Importantly, it discriminates treated from untreated patients, uses features commonly found in such biopsies, and is utilizable by pathologists after minimal training. These data provide rationales and a method to evaluate esophageal biopsies for features in addition to PEC.


Subject(s)
Biopsy/statistics & numerical data , Eosinophilic Esophagitis/diagnosis , Eosinophils , Leukocyte Count/methods , Severity of Illness Index , Area Under Curve , Biopsy/methods , Child , Esophagus/pathology , Female , Humans , Logistic Models , Male , Prospective Studies , Reproducibility of Results
12.
Mucosal Immunol ; 10(1): 194-204, 2017 01.
Article in English | MEDLINE | ID: mdl-27049063

ABSTRACT

Eosinophils contribute to type II immune responses in helminth infections and allergic diseases; however, their influence on intracellular pathogens is less clear. We previously reported that CCR2-/- mice exposed to the intracellular fungal pathogen Histoplasma capsulatum exhibit dampened immunity caused by an early exaggerated interleukin (IL)-4 response. We sought to identify the cellular source promulgating IL-4 in infected mutant animals. Eosinophils were the principal instigators of non-protective IL-4 and depleting this granulocyte population improved fungal clearance in CCR2-/- animals. The deleterious impact of eosinophilia on mycosis was also recapitulated in transgenic animals overexpressing eosinophils. Mechanistic examination of IL-4 induction revealed that phagocytosis of H. capsulatum via the pattern recognition receptor complement receptor (CR) 3 triggered the heightened IL-4 response in murine eosinophils. This phenomenon was conserved in human eosinophils; exposure of cells to the fungal pathogen elicited a robust IL-4 response. Thus, our findings elucidate a detrimental attribute of eosinophil biology in fungal infections that could potentially trigger a collapse in host defenses by instigating type II immunity.


Subject(s)
Eosinophils/immunology , Histoplasma/immunology , Histoplasmosis/immunology , Interleukin-4/metabolism , Receptors, CCR2/metabolism , Animals , Antigens, Fungal/immunology , Cells, Cultured , Eosinophils/microbiology , Humans , Immune Evasion , Immunity, Innate , Macrophage-1 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Receptors, CCR2/genetics
14.
Allergy ; 71(5): 611-20, 2016 05.
Article in English | MEDLINE | ID: mdl-26799684

ABSTRACT

Eosinophilic esophagitis (EoE) is a chronic disease characterized clinically by symptoms of esophageal dysfunction and histologically by eosinophil-predominant inflammation. EoE is frequently associated with concomitant atopic diseases and immunoglobulin E (IgE) sensitization to food allergens in children as well as to aeroallergens and cross-reactive plant allergen components in adults. Patients with EoE respond well to elemental and empirical food elimination diets. Recent research has, however, indicated that the pathogenesis of EoE is distinct from IgE-mediated food allergy. In this review, we discuss the individual roles of epithelial barrier defects, dysregulated innate and adaptive immune responses, and of microbiota in the pathogenesis of EoE. Although food has been recognized as a trigger factor of EoE, the mechanism by which it initiates or facilitates eosinophilic inflammation appears to be largely independent of IgE and needs to be further investigated. Understanding the pathogenic role of food in EoE is a prerequisite for the development of specific diagnostic tools and targeted therapeutic procedures.


Subject(s)
Eosinophilic Esophagitis/diagnosis , Eosinophilic Esophagitis/etiology , Food Hypersensitivity/diagnosis , Food Hypersensitivity/etiology , Allergens/immunology , Anti-Asthmatic Agents/therapeutic use , Eosinophilic Esophagitis/drug therapy , Eosinophilic Esophagitis/metabolism , Epithelium/immunology , Epithelium/metabolism , Epithelium/pathology , Food/adverse effects , Food Hypersensitivity/metabolism , Humans , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/pathology , Immunity, Innate , Immunoglobulin E/immunology , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Omalizumab/therapeutic use , Skin/immunology , Skin/metabolism , Skin/pathology , Th2 Cells/immunology , Th2 Cells/metabolism , Treatment Outcome
15.
Mucosal Immunol ; 9(3): 744-56, 2016 05.
Article in English | MEDLINE | ID: mdl-26462420

ABSTRACT

Eosinophilic esophagitis (EoE) is an allergic inflammatory disease of the esophagus featuring increased esophageal interleukin-13 (IL-13) levels and impaired barrier function. Herein, we investigated leucine-rich repeat-containing protein 31 (LRRC31) in human EoE esophageal tissue and IL-13-treated esophageal epithelial cells. LRRC31 had basal mRNA expression in colonic and airway mucosal epithelium. Esophageal LRRC31 mRNA and protein increased in active EoE and strongly correlated with esophageal eosinophilia and IL13 and CCL26 (chemokine (C-C motif) ligand 26) mRNA expression. IL-13 treatment increased LRRC31 mRNA and protein in air-liquid interface-differentiated esophageal epithelial cells (EPC2s). At baseline, differentiated LRRC31-overexpressing EPC2s had increased barrier function (1.9-fold increase in transepithelial electrical resistance (P<0.05) and 2.8-fold decrease in paracellular flux (P<0.05)). RNA sequencing analysis of differentiated LRRC31-overexpressing EPC2s identified 38 dysregulated genes (P<0.05), including five kallikrein (KLK) serine proteases. Notably, differentiated LRRC31-overexpressing EPC2s had decreased KLK expression and activity, whereas IL-13-treated, differentiated LRRC31 gene-silenced EPC2s had increased KLK expression and suprabasal epithelial detachment. We identified similarly dysregulated KLK expression in the esophagus of patients with active EoE and in IL-13-treated esophageal epithelial cells. We propose that LRRC31 is induced by IL-13 and modulates epithelial barrier function, potentially through KLK regulation.


Subject(s)
Eosinophilic Esophagitis/immunology , Epithelium/metabolism , Esophagus/immunology , Interleukin-13/metabolism , Kallikreins/metabolism , Nuclear Proteins/metabolism , Proteins/metabolism , Adult , Biological Transport , Cell Differentiation , Cell Line , Dextrans/pharmacokinetics , Electric Impedance , Epithelium/pathology , Esophagus/pathology , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/pharmacokinetics , Gene Expression Regulation , Humans , Interleukin-13/genetics , Kallikreins/genetics , Leucine-Rich Repeat Proteins , Nuclear Proteins/genetics , Proteins/genetics , RNA, Small Interfering/genetics
16.
Mucosal Immunol ; 8(3): 464-75, 2015 May.
Article in English | MEDLINE | ID: mdl-25807184

ABSTRACT

Eosinophils, multifunctional cells that contribute to both innate and adaptive immunity, are involved in the initiation, propagation, and resolution of immune responses, including tissue repair. They achieve this multifunctionality by expression of a diverse set of activation receptors, including those that directly recognize pathogens and opsonized targets, and by their ability to store and release preformed cytotoxic mediators that participate in host defense, to produce a variety of de novo pleotropic mediators and cytokines, and to interact directly and indirectly with diverse cell types, including adaptive and innate immunocytes and structural cells. Herein, we review the basic biology of eosinophils and then focus on new emerging concepts about their role in mucosal immune homeostasis, particularly maintenance of intestinal IgA. We review emerging data about their development and regulation and describe new concepts concerning mucosal eosinophilic diseases. We describe recently developed therapeutic strategies to modify eosinophil levels and function and provide collective insight about the beneficial and detrimental functions of these enigmatic cells.


Subject(s)
Asthma/immunology , Enteritis/immunology , Eosinophilia/immunology , Eosinophilic Esophagitis/immunology , Eosinophils/immunology , Gastritis/immunology , Immunity, Mucosal , Inflammatory Bowel Diseases/immunology , Adaptive Immunity , Animals , Antibodies, Monoclonal/therapeutic use , Asthma/drug therapy , Asthma/pathology , Cytokines/genetics , Cytokines/immunology , Enteritis/drug therapy , Enteritis/pathology , Eosinophilia/drug therapy , Eosinophilia/pathology , Eosinophilic Esophagitis/drug therapy , Eosinophilic Esophagitis/pathology , Eosinophils/drug effects , Eosinophils/pathology , Gastritis/drug therapy , Gastritis/pathology , Gene Expression , Homeostasis/drug effects , Homeostasis/immunology , Humans , Immunity, Innate , Immunologic Factors/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology
17.
Clin Pharmacol Ther ; 97(5): 502-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25676488

ABSTRACT

Although randomized trials provide the most reliable evidence of a drug's safety and efficacy, there are situations where randomized trials are not possible or ethical. In this article we discuss when and how single-arm trials can be used to support full approval of oncology drugs. These include situations in which an unprecedented effect on tumor response is observed in a setting of high unmet medical need, clinical trial patients have been well characterized, enabling a target population to be clearly defined, experience exists in a sufficient number of patients to allow adequate assessment of the risk:benefit relationship, and a proper historical context can be provided for analysis. We also discuss how response rates might be considered predictive of long-term outcomes or clinically meaningful in and of themselves in certain contexts.


Subject(s)
Antineoplastic Agents/therapeutic use , Clinical Trials as Topic/methods , Evidence-Based Medicine/methods , Medical Oncology/methods , Neoplasms/drug therapy , Research Design , Antineoplastic Agents/adverse effects , Clinical Trials as Topic/standards , Drug Approval , Endpoint Determination , Evidence-Based Medicine/standards , Humans , Medical Oncology/standards , Practice Guidelines as Topic , Research Design/standards , Risk Assessment , Risk Factors , Treatment Outcome
18.
Mucosal Immunol ; 8(4): 930-42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25563499

ABSTRACT

Eosinophils are multifunctional leukocytes that reside in the gastrointestinal (GI) lamina propria, where their basal function remains largely unexplored. In this study, by examining mice with a selective deficiency of systemic eosinophils (by lineage ablation) or GI eosinophils (eotaxin-1/2 double deficient or CC chemokine receptor 3 deficient), we show that eosinophils support immunoglobulin A (IgA) class switching, maintain intestinal mucus secretions, affect intestinal microbial composition, and promote the development of Peyer's patches. Eosinophil-deficient mice showed reduced expression of mediators of secretory IgA production, including intestinal interleukin 1ß (IL-1ß), inducible nitric oxide synthase, lymphotoxin (LT) α, and LT-ß, and reduced levels of retinoic acid-related orphan receptor gamma t-positive (ROR-γt(+)) innate lymphoid cells (ILCs), while maintaining normal levels of APRIL (a proliferation-inducing ligand), BAFF (B cell-activating factor of the tumor necrosis factor family), and TGF-ß (transforming growth factor ß). GI eosinophils expressed a relatively high level of IL-1ß, and IL-1ß-deficient mice manifested the altered gene expression profiles observed in eosinophil-deficient mice and decreased levels of IgA(+) cells and ROR-γt(+) ILCs. On the basis of these collective data, we propose that eosinophils are required for homeostatic intestinal immune responses including IgA production and that their affect is mediated via IL-1ß in the small intestine.


Subject(s)
Eosinophils/immunology , Eosinophils/metabolism , Homeostasis , Immunoglobulin A/biosynthesis , Interleukin-1beta/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Adoptive Transfer , Animals , Cell Count , Gastrointestinal Microbiome , Gene Expression , Immune Tolerance , Immunoglobulin A, Secretory/biosynthesis , Interleukin-1beta/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestine, Small/microbiology , Lymphotoxin-alpha/genetics , Lymphotoxin-beta/genetics , Mice , Mice, Knockout , Mucus/metabolism , Peyer's Patches/immunology , Peyer's Patches/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism
19.
Leukemia ; 29(4): 828-38, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25322685

ABSTRACT

In acute myeloid leukemia (AML), several signaling pathways such as the phosphatidylinositol-3-kinase/AKT and the mammalian target of rapamycin (PI3K/AKT/mTOR) pathway are deregulated and constitutively activated as a consequence of genetic and cytogenetic abnormalities. We tested the effectiveness of PI3K/AKT/mTOR-targeting therapies and tried to identify alterations that associate with treatment sensitivity. By analyzing primary samples and cell lines, we observed a wide range of cytotoxic activity for inhibition of AKT (MK-2206), mTORC1 (rapamycin) and PI3K/mTORC1/2 (BEZ-235) with a high sensitivity of cells carrying an MLL rearrangement. In vivo PI3K/mTOR inhibition delayed tumor progression, reduced tumor load and prolonged survival in an MLL-AF9(+)/FLT3-ITD(+) xenograft mouse model. By performing targeted amplicon sequencing in 38 MLL-AF9(+) and 125 cytogenetically normal AML patient samples, we found a high additional mutation rate for genes involved in growth factor signaling in 79% of all MLL-AF9(+) samples, which could lead to a possible benefit of this cohort. PI3K/mTOR inhibition for 24 h led to the cross-activation of the ERK pathway. Further in vitro studies combining PI3K/mTOR and ERK pathway inhibition revealed highly synergistic effects in apoptosis assays. Our data implicate a possible therapeutic benefit of PI3K/mTOR inhibition in the MLL-mutated subgroup. Inhibiting rescue pathways could improve the therapeutic efficacy of PI3K-targeted therapies in AML.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Drug Synergism , Gene Rearrangement , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Imidazoles/pharmacology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Quinolines/pharmacology , Signal Transduction , Sirolimus/pharmacology , Survival Analysis , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
20.
Mucosal Immunol ; 8(4): 785-98, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25389033

ABSTRACT

Although interleukin (IL)-13 and neurotrophins are functionally important for the pathogenesis of immune responses, the interaction of these pathways has not been explored. Herein, by interrogating IL-13-induced responses in human epithelial cells we show that neurotrophic tyrosine kinase receptor, type 1 (NTRK1), a cognate, high-affinity receptor for nerve growth factor (NGF), is an early transcriptional IL-13 target. Induction of NTRK1 was accompanied by accumulation of activating epigenetic marks in the promoter; transcriptional and epigenetic changes were signal transducer and activator of transcription 6 dependent. Using eosinophilic esophagitis as a model for human allergic inflammation, we found that NTRK1 was increased in inflamed tissue and dynamically expressed as a function of disease activity and that the downstream mediator of NTRK1 signaling early growth response 1 protein was elevated in allergic inflammatory tissue compared with control tissue. Unlike NTRK1, its ligand NGF was constitutively expressed in control and disease states, indicating that IL-13-stimulated NTRK1 induction is a limiting factor in pathway activation. In epithelial cells, NGF and IL-13 synergistically induced several target genes, including chemokine (C-C motif) ligand 26 (eotaxin-3). In summary, we have demonstrated that IL-13 confers epithelial cell responsiveness to NGF by regulating NTRK1 levels by a transcriptional and epigenetic mechanism and that this process likely contributes to allergic inflammation.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Hypersensitivity/genetics , Hypersensitivity/metabolism , Interleukin-13/metabolism , Receptor, trkA/genetics , Transcription, Genetic , Cluster Analysis , Early Growth Response Protein 1/metabolism , Eosinophilic Esophagitis/genetics , Eosinophilic Esophagitis/metabolism , Eosinophilic Esophagitis/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Silencing , Humans , Interleukin-13/pharmacology , Nerve Growth Factor/pharmacology , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...