Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
1.
Cancer Lett ; 523: 182-194, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34627950

ABSTRACT

Rab1A overexpression has been observed in several cancer types, however, its significance and the underlying mechanisms in non-small cell lung cancer (NSCLC) remain largely unexplored. This study demonstrated that Rab1A overexpression in NSCLC was significantly correlated to short survival and metastasis. Rab1A overexpression promoted cancer cell migration, invasion, and metastasis both in vitro and in vivo, by activating JAK1/STAT6 signaling through stabilizing IL-4Rα protein. Strikingly, high Rab1A level was associated with sensitivity to JAK1 inhibitor, and Rab1A overexpression rendered cancer cells vulnerable to JAK1-targeted agents. JAK1 inhibitor, Itacitinib adipate, dramatically inhibited high Rab1A NSCLC metastasis, in both cell line and patient derived xenograft models. Collectively, these findings demonstrated that Rab1A plays a critical role in the aggressive properties of NSCLC, revealing a unique mechanism by which it promotes metastasis. In addition, we found that Rab1A is a determinant of JAK1 inhibitor sensitivity, which could be explored for improving JAK1-targeted cancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Janus Kinase 1/antagonists & inhibitors , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Receptors, Interleukin-4/physiology , STAT6 Transcription Factor/physiology , rab1 GTP-Binding Proteins/physiology , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement , Humans , Janus Kinase 1/physiology , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Metastasis , Xenograft Model Antitumor Assays
2.
JCI Insight ; 5(4)2020 02 27.
Article in English | MEDLINE | ID: mdl-32102987

ABSTRACT

IL-4 is a pleiotropic antiinflammatory cytokine, which can be neuroprotective after nervous system injury. The beneficial actions of IL-4 are thought to result from the blunting of action of inflammatory mediators, such as proinflammatory cytokines. Here, we demonstrate that IL-4 induces M2 macrophages to continuously produce opioid peptides and ameliorate pain. IL-4 application at injured nerves in mice shifted F4/80+ macrophages from the proinflammatory M1 to the antiinflammatory M2 phenotype, which synthesized opioid peptides (Met-enkephalin, ß-endorphin, and dynorphin A 1-17). These effects were accompanied by a long-lasting attenuation of neuropathy-induced mechanical hypersensitivity, beyond the IL-4 treatment. This IL-4-induced analgesia was decreased by opioid peptide antibodies and opioid receptor (δ, µ, κ) antagonists applied at injured nerves, which confirms the involvement of the local opioid system. The participation of M2 macrophages was supported by analgesia in recipient mice injected at injured nerves with F4/80+ macrophages from IL-4-treated donors. Together, IL-4-induced M2 macrophages at injured nerves produced opioid peptides, which activated peripheral opioid receptors to diminish pain. Fostering the opioid-mediated actions of intrinsic M2 macrophages may be a strategy to tackle pathological pain.


Subject(s)
Analgesia , Interleukin-4/pharmacology , Macrophages/drug effects , Opioid Peptides/biosynthesis , Animals , Hot Temperature , Interleukin-4/therapeutic use , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Neuralgia/drug therapy , Opioid Peptides/physiology , Reaction Time/drug effects , Receptors, Interleukin-4/antagonists & inhibitors , Receptors, Interleukin-4/physiology
3.
J Immunol ; 203(9): 2532-2544, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31548328

ABSTRACT

Dual oxidase 2 (DUOX2) generates H2O2 that plays a critical role in both host defense and chronic inflammation. Previously, we demonstrated that the proinflammatory mediators IFN-γ and LPS enhance expression of DUOX2 and its maturation factor DUOXA2 through STAT1- and NF-κB‒mediated signaling in human pancreatic cancer cells. Using a panel of colon and pancreatic cancer cell lines, we now report the induction of DUOX2/DUOXA2 mRNA and protein expression by the TH2 cytokine IL-4. IL-4 activated STAT6 signaling that, when silenced, significantly decreased induction of DUOX2. Furthermore, the TH17 cytokine IL-17A combined synergistically with IL-4 to increase DUOX2 expression in both colon and pancreatic cancer cells mediated, at least in part, by signaling through NF-κB. The upregulation of DUOX2 was associated with a significant increase in the production of extracellular H2O2 and DNA damage-as indicated by the accumulation of 8-oxo-dG and γH2AX-which was suppressed by the NADPH oxidase inhibitor diphenylene iodonium and a DUOX2-specific small interfering RNA. The clinical relevance of these experiments is suggested by immunohistochemical, microarray, and quantitative RT-PCR studies of human colon and pancreatic tumors demonstrating significantly higher DUOX2, IL-4R, and IL-17RA expression in tumors than in adjacent normal tissues; in pancreatic adenocarcinoma, increased DUOX2 expression is adversely associated with overall patient survival. These data suggest a functional association between DUOX2-mediated H2O2 production and induced DNA damage in gastrointestinal malignancies.


Subject(s)
Colonic Neoplasms/metabolism , DNA Damage , Dual Oxidases/genetics , Hydrogen Peroxide/metabolism , Interleukin-17/pharmacology , Interleukin-4/pharmacology , Pancreatic Neoplasms/metabolism , Cell Line, Tumor , Colonic Neoplasms/pathology , Humans , NF-kappa B/physiology , Oxidation-Reduction , Pancreatic Neoplasms/pathology , Receptors, Interleukin-4/physiology , STAT6 Transcription Factor/physiology , Signal Transduction , Up-Regulation
4.
J Exp Med ; 214(8): 2205-2216, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28694386

ABSTRACT

In the thymus, stromal microenvironments support a developmental program that generates mature T cells ready for thymic exit. The cellular and molecular specialization within thymic stromal cells that enables their regulation of specific stages of thymocyte development is poorly understood. Here, we show the thymic microenvironment expresses the type 2 IL-4R complex and is functionally responsive to its known ligands, IL-4 and IL-13. Absence of IL-4Rα limits thymocyte emigration, leading to an intrathymic accumulation of mature thymocytes within medullary perivascular spaces and reduced numbers of recent thymic emigrants. Thymus transplantation shows this requirement maps to IL-4Rα expression by stromal cells, and we provide evidence that it regulates thymic exit via a process distinct from S1P-mediated migration. Finally, we reveal a cellular mechanism by which IL-4+IL-13+ invariant NKT cells are necessary for IL-4Rα signaling that regulates thymic exit. Collectively, we define a new axis for thymic emigration involving stimulation of the thymic microenvironment via type 2 cytokines from innate T cells.


Subject(s)
Receptors, Interleukin-4/physiology , Thymus Gland/physiology , Animals , Cell Movement/physiology , Interleukin-13/physiology , Interleukin-4/physiology , Mice , Mice, Knockout , Natural Killer T-Cells/physiology , Signal Transduction/physiology , Thymocytes/physiology , Thymus Gland/transplantation
5.
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
6.
Clin Exp Metastasis ; 32(8): 847-56, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26385103

ABSTRACT

While progress has been made in treating primary epithelial tumors, metastatic tumors remain largely incurable and still account for 85-90 % of all cancer-related deaths. Interleukin-4 (IL4), a Th2 cytokine, and the IL4/IL4 receptor (IL4R) interaction have well defined roles in the immune system. Yet, IL4 receptors are over-expressed by many epithelial cancers and could be a promising target for metastatic tumor therapy. The IL4/IL4R signaling axis is a strong promoter of pro-metastatic phenotypes in epithelial cancer cells including enhanced migration, invasion, survival, and proliferation. The promotion of breast cancer growth specifically is also supported in part by IL4-induced glutamine metabolism, and we have shown that IL4 is also capable of inducing glucose metabolism in breast cancer cells. Importantly, there are several types of FDA approved medications for use in asthma patients that inhibit the IL4/IL4R signaling axis. However, these approved medications inhibit both the type I IL4 receptor found on immune cells, and the type II IL4 receptor that is predominantly expressed by some non-hematopoietic cells including epithelial cancer cells. This article reviews existing therapies targeting IL4, IL4R, or IL4/IL4R signaling, and recent findings guiding the creation of novel therapies that specifically inhibit the type II IL4R, while taking into consideration effects on immune cells within the tumor microenvironment. Some of these therapies are currently in clinical trials for cancer patients, and may be exploitable for the treatment of metastatic disease.


Subject(s)
Interleukin-4/antagonists & inhibitors , Neoplasms, Glandular and Epithelial/drug therapy , Receptors, Interleukin-4/antagonists & inhibitors , Humans , Interleukin-4/physiology , Janus Kinase 1/antagonists & inhibitors , Neoplasms, Glandular and Epithelial/pathology , Receptors, Interleukin-4/physiology , STAT6 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Tumor Microenvironment
7.
Nat Med ; 21(9): 1010-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26280119

ABSTRACT

T cell stimulation via glucocorticoid-induced tumor necrosis factor receptor (TNFR)-related protein (GITR) elicits antitumor activity in various tumor models; however, the underlying mechanism of action remains unclear. Here we demonstrate a crucial role for interleukin (IL)-9 in antitumor immunity generated by the GITR agonistic antibody DTA-1. IL-4 receptor knockout (Il4ra(-/-)) mice, which have reduced expression of IL-9, were resistant to tumor growth inhibition by DTA-1. Notably, neutralization of IL-9 considerably impaired tumor rejection induced by DTA-1. In particular, DTA-1-induced IL-9 promoted tumor-specific cytotoxic T lymphocyte (CTL) responses by enhancing the function of dendritic cells in vivo. Furthermore, GITR signaling enhanced the differentiation of IL-9-producing CD4(+) T-helper (TH9) cells in a TNFR-associated factor 6 (TRAF6)- and NF-κB-dependent manner and inhibited the generation of induced regulatory T cells in vitro. Our findings demonstrate that GITR co-stimulation mediates antitumor immunity by promoting TH9 cell differentiation and enhancing CTL responses and thus provide a mechanism of action for GITR agonist-mediated cancer immunotherapies.


Subject(s)
Glucocorticoid-Induced TNFR-Related Protein/physiology , Glucocorticoids/pharmacology , Interleukin-9/physiology , Neoplasms, Experimental/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Differentiation , Dendritic Cells/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-kappa B/physiology , Neoplasms, Experimental/drug therapy , Receptors, Interleukin-4/physiology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , TNF Receptor-Associated Factor 6/physiology
9.
Am J Respir Crit Care Med ; 189(1): 16-29, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24251647

ABSTRACT

RATIONALE: IL-4Rα, the common receptor component for IL-4 and IL-13, plays a critical role in IL-4- and IL-13-mediated signaling pathways that regulate airway inflammation and remodeling. However, the regulatory mechanisms underlying IL-4Rα turnover and its signal termination remain elusive. OBJECTIVES: To evaluate the role of STUB1 (STIP1 homology and U-Box containing protein 1) in regulating IL-4R signaling in airway inflammation. METHODS: The roles of STUB1 in IL-4Rα degradation and its signaling were investigated by immunoblot, immunoprecipitation, and flow cytometry. The involvement of STUB1 in airway inflammation was determined in vivo by measuring lung inflammatory cells infiltration, mucus production, serum lgE levels, and alveolar macrophage M2 activation in STUB1(-/-) mice. STUB1 expression was evaluated in airway epithelium of patients with asthma and lung tissues of subjects with chronic obstructive pulmonary disease. MEASUREMENTS AND MAIN RESULTS: STUB1 interacted with IL-4Rα and targeted it for ubiquitination-mediated proteasomal degradation, terminating IL-4 or IL-13 signaling. STUB1 knockout cells showed increased levels of IL-4Rα and sustained STAT6 activation, whereas STUB1 overexpression reduced IL-4Rα levels. Mice deficient in STUB1 had spontaneous airway inflammation, alternative M2 activation of alveolar macrophage, and increased serum IgE. STUB1 levels were increased in airways of subjects with asthma or chronic obstructive pulmonary disease, suggesting that up-regulation of STUB1 might be an important feedback mechanism to dampen IL-4R signaling in airway inflammation. CONCLUSIONS: Our study identified a previously uncharacterized role for STUB1 in regulating IL-4R signaling, which might provide a new strategy for attenuating airway inflammation.


Subject(s)
Pneumonia/physiopathology , Receptors, Interleukin-4/physiology , Signal Transduction/physiology , Ubiquitin-Protein Ligases/physiology , Adult , Animals , Asthma/physiopathology , Child , Down-Regulation/physiology , Female , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , Macrophage Activation/physiology , Male , Mice , Mice, Knockout , Proteasome Endopeptidase Complex/physiology , Pulmonary Disease, Chronic Obstructive/physiopathology , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/physiology , STAT6 Transcription Factor/physiology , Ubiquitin/physiology
10.
Cancer Res ; 72(24): 6338-43, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23222300

ABSTRACT

The Th2 cytokines interleukin (IL)-4 and -13 are acknowledged regulators of lymphocyte proliferation and activation. They have also been well studied in the regulation of various myeloid-derived populations in tumor biology. It has become clear, however, that both cytokines can have direct effects on epithelial tumor cells expressing appropriate receptors. Changes in tumor proliferation, survival, and metastatic capability have all been ascribed to IL-4 and/or IL-13 action. Here, we evaluate the evidence to support direct tumor-promoting roles of these cytokines. We also identify the questions that should be addressed before proceeding with therapeutic approaches based on neutralization of IL-4 or IL-13 pathways.


Subject(s)
Cytokines/pharmacology , Interleukin-13/physiology , Interleukin-4/physiology , Neoplasms, Glandular and Epithelial/immunology , Neoplasms, Glandular and Epithelial/pathology , Animals , Cytokines/physiology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Interleukin-13/metabolism , Interleukin-13/pharmacology , Interleukin-4/metabolism , Interleukin-4/pharmacology , Models, Biological , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/metabolism , Receptors, Interleukin-13/genetics , Receptors, Interleukin-13/metabolism , Receptors, Interleukin-13/physiology , Receptors, Interleukin-4/genetics , Receptors, Interleukin-4/metabolism , Receptors, Interleukin-4/physiology , Stimulation, Chemical
11.
PLoS One ; 7(6): e39673, 2012.
Article in English | MEDLINE | ID: mdl-22761864

ABSTRACT

BACKGROUND: Previous work from our laboratory demonstrated that IL-4Rα expression on a myeloid cell type was responsible for enhancement of Th2-driven eosinophilic inflammation in a mouse model of allergic lung inflammation. Subsequently, we have shown that IL-4 signaling through type I IL-4 receptors on monocytes/macrophages strongly induced activation of the IRS-2 pathway and a subset of genes characteristic of alternatively activated macrophages. The direct effect(s) of IL-4 and IL-13 on mouse eosinophils are not clear. The goal of this study was determine the effect of IL-4 and IL-13 on mouse eosinophil function. METHODS: Standard Transwell chemotaxis assay was used to assay migration of mouse eosinophils and signal transduction was assessed by Western blotting. RESULTS: Here we determined that (i) mouse eosinophils express both type I and type II IL-4 receptors, (ii) in contrast to human eosinophils, mouse eosinophils do not chemotax to IL-4 or IL-13 although (iii) pre-treatment with IL-4 but not IL-13 enhanced migration to eotaxin-1. This IL-4-mediated enhancement was dependent on type I IL-4 receptor expression: γC-deficient eosinophils did not show enhancement of migratory capacity when pre-treated with IL-4. In addition, mouse eosinophils responded to IL-4 with the robust tyrosine phosphorylation of STAT6 and IRS-2, while IL-13-induced responses were considerably weaker. CONCLUSIONS: The presence of IL-4 in combination with eotaxin-1 in the allergic inflammatory milieu could potentiate infiltration of eosinophils into the lungs. Therapies that block IL-4 and chemokine receptors on eosinophils might be more effective clinically in reducing eosinophilic lung inflammation.


Subject(s)
Chemokine CCL11/metabolism , Eosinophils/cytology , Interleukin-4/metabolism , Receptors, Interleukin-4/metabolism , Animals , Base Sequence , Blotting, Western , Chemokine CCL11/physiology , DNA Primers , In Vitro Techniques , Interleukin-4/physiology , Mice , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-4/physiology , Signal Transduction
12.
J Leukoc Biol ; 92(4): 753-64, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22782966

ABSTRACT

Studies of IL-4 have revealed a wealth of information on the diverse roles of this cytokine in homeostatic regulation and disease pathogenesis. Recent data suggest that instead of simple linear regulatory pathways, IL-4 drives regulation that is full of alternatives. In addition to the well-known dichotomous regulation of Th cell differentiation by IL-4, this cytokine is engaged in several other alternative pathways. Its own production involves alternative mRNA splicing, yielding at least two functional isoforms: full-length IL-4, encoded by the IL-4 gene exons 1-4, and IL-4δ2, encoded by exons 1, 3, and 4. The functional effects of these two isoforms are in some ways similar but in other ways quite distinct. When binding to the surface of target cells, IL-4 may differentially engage two different types of receptors. By acting on macrophages, a cell type critically involved in inflammation, IL-4 induces the so-called alternative macrophage activation. In this review, recent advances in understanding these three IL-4-related branch points--alternative splicing of IL-4, differential receptor engagement by IL-4, and differential regulation of macrophage activation by IL-4--are summarized in light of their contributions to inflammation.


Subject(s)
Inflammation/etiology , Interleukin-4/physiology , Animals , Asthma/etiology , Humans , Interleukin-4/genetics , Macrophage Activation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Receptors, Interleukin-13/chemistry , Receptors, Interleukin-13/physiology , Receptors, Interleukin-4/chemistry , Receptors, Interleukin-4/physiology , Scleroderma, Systemic/etiology , Signal Transduction , Tuberculosis/etiology
13.
J Immunol ; 188(11): 5571-84, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22547699

ABSTRACT

The roles of IL-4 and IL-4Rα in Th2-mediated immunity have been well characterized in humans and other mammals. In contrast, few reports have been documented in ancient vertebrates. Several putative IL-4- and IL-4Rα-like molecules were identified recently from a few fish species, providing preliminary insight into the occurrence of Th2-type immunity in teleosts. However, functional determination still is required to address this hypothesis. To this end, these two molecules were characterized functionally in zebrafish (Danio rerio). Besides the identification of a full-length IL-4Rα molecule and an isoform lacking most of the cytoplasmic region as predicted previously, two novel alternatively spliced soluble variants with the extracellular domain only also were identified. Zebrafish IL-4Rα (DrIL-4Rα) shared overall conserved structural features of the IL-4Rα family. Immunofluorescence staining showed that DrIL-4Rα distributed on B cells. In vitro binding assays demonstrated that zebrafish IL-4 (DrIL-4) can bind specifically to DrIL-4Rα. In vivo administration of DrIL-4 significantly upregulated B cell proliferation and Ab production. These DrIL-4-elicited immune responses were downregulated by the administration of zebrafish soluble IL-4Rα or by DrIL-4Rα blockade using anti-DrIL-4Rα Abs. In addition, Th2-related cytokines or transcription factors were upregulated by DrIL-4. The DrIL-4-DrIL-4Rα interaction promoted CD40 expression on B cells and enhanced the CD154-CD40 costimulatory response, both of which are crucial for the initiation of Th2-type immunity. To our knowledge, this is the first report showing that a possible Th2-mediated regulatory mechanism may have appeared before the divergence of teleosts and mammals. These results add greater insight into the evolutionary history of adaptive immunity.


Subject(s)
Adaptive Immunity/immunology , Interleukin-4/physiology , Receptors, Interleukin-4/physiology , Th2 Cells/immunology , Zebrafish/immunology , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Biological Evolution , Birds , CD40 Antigens/physiology , CD40 Ligand/physiology , Cattle , Chickens , Conserved Sequence/immunology , Dogs , Horses , Humans , Interleukin-4/chemistry , Interleukin-4/metabolism , Macaca mulatta , Male , Mice , Mice, Inbred BALB C , Pan troglodytes , Rabbits , Rats , Receptors, Interleukin-4/chemistry , Receptors, Interleukin-4/metabolism , Signal Transduction/immunology
14.
Clin Cancer Res ; 17(9): 2757-66, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21536546

ABSTRACT

PURPOSE: Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in childhood. The alveolar subtype of rhabdomyosarcoma (ARMS) is a paradigm for refractory and incurable solid tumors because more than half of the children at diagnosis have either regional lymph node or distant metastases. These studies follow our previous observation that Interleukin-4 receptor α (IL-4Rα) is upregulated in both human and murine ARMS, and that the IL-4R signaling pathway may be a target for abrogating tumor progression. EXPERIMENTAL DESIGN: By in vitro biochemical and cell biology studies as well as preclinical studies using a genetically engineered mouse model, we evaluated the role of IL-4 and IL-13 in IL-4R-mediated mitogenesis, myodifferentiation, and tumor progression. RESULTS: IL-4 and IL-13 ligands accelerated tumor cell growth and activated STAT6, Akt, or MAPK signaling pathways in the human RMS cell lines, RD and Rh30, as well as in mouse primary ARMS cell cultures. IL-4 and IL-13 treatment also decreased protein expression of myogenic differentiation factors MyoD and Myogenin, indicating a loss of muscle differentiation. Using a genetically engineered mouse model of ARMS, we have shown that inhibition of IL-4R signaling pathway with a neutralizing antibody has a profound effect on the frequency of lymph node and pulmonary metastases, resulting in significant survival extension in vivo. CONCLUSIONS: Our results indicate that an IL-4R-dependent signaling pathway regulates tumor cell progression in RMS, and inhibition of this pathway could be a promising adjuvant therapeutic approach.


Subject(s)
Cell Dedifferentiation/genetics , Cell Transformation, Neoplastic/genetics , Muscle Neoplasms/genetics , Receptors, Interleukin-4/physiology , Rhabdomyosarcoma/genetics , Animals , Cell Transformation, Neoplastic/chemically induced , Cells, Cultured , Disease Models, Animal , Genes, p53 , Humans , Mice , Mice, Transgenic , Mitogens , Muscle Neoplasms/pathology , Myogenic Regulatory Factors/genetics , Neoplasm Metastasis , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Receptors, Interleukin-4/genetics , Rhabdomyosarcoma/pathology , Signal Transduction/genetics , Signal Transduction/physiology
15.
J Exp Med ; 208(4): 853-67, 2011 Apr 11.
Article in English | MEDLINE | ID: mdl-21464224

ABSTRACT

Production of the cytokines IL-4 and IL-13 is increased in both human asthma and mouse asthma models, and Stat6 activation by the common IL-4/IL-13R drives most mouse model pathophysiology, including airway hyperresponsiveness (AHR). However, the precise cellular mechanisms through which IL-4Rα induces AHR remain unclear. Overzealous bronchial smooth muscle constriction is thought to underlie AHR in human asthma, but the smooth muscle contribution to AHR has never been directly assessed. Furthermore, differences in mouse versus human airway anatomy and observations that selective IL-13 stimulation of Stat6 in airway epithelium induces murine AHR raise questions about the importance of direct IL-4R effects on smooth muscle in murine asthma models and the relevance of these models to human asthma. Using transgenic mice in which smooth muscle is the only cell type that expresses or fails to express IL-4Rα, we demonstrate that direct smooth muscle activation by IL-4, IL-13, or allergen is sufficient but not necessary to induce AHR. Five genes known to promote smooth muscle migration, proliferation, and contractility are activated by IL-13 in smooth muscle in vivo. These observations demonstrate that IL-4Rα promotes AHR through multiple mechanisms and provide a model for testing smooth muscle-directed asthma therapeutics.


Subject(s)
Bronchial Hyperreactivity/etiology , Muscle, Smooth/physiology , Receptors, Interleukin-4/physiology , Allergens/immunology , Animals , Female , Gene Expression Regulation , Interleukin-13/pharmacology , Interleukin-4/pharmacology , Mice , Mice, Inbred BALB C , Muscle, Smooth/drug effects , Receptors, Interleukin-4/genetics
16.
Am J Hum Biol ; 23(1): 22-8, 2011.
Article in English | MEDLINE | ID: mdl-21140465

ABSTRACT

OBJECTIVES: Von Economo neurons (VENs) are defined by their thin, elongated cell body and long dendrites projecting from apical and basal ends. These distinctive neurons are mostly present in anterior cingulate (ACC) and fronto-insular (FI) cortex, with particularly high densities in cetaceans, elephants, and hominoid primates (i.e., humans and apes). This distribution suggests that VENs contribute to specializations of neural circuits in species that share both large brain size and complex social cognition, possibly representing an adaptation to rapidly relay socially-relevant information over long distances across the brain. Recent evidence indicates that unique patterns of protein expression may also characterize VENs, particularly involving molecules that are known to regulate gut and immune function. METHODS: In this study, we used quantitative stereologic methods to examine the expression of three such proteins that are localized in VENs-activating-transcription factor 3 (ATF3), interleukin 4 receptor (IL4Rα), and neuromedin B (NMB). We quantified immunoreactivity against these proteins in different morphological classes of ACC layer V neurons of hominoids. RESULTS: Among the different neuron types analyzed (pyramidal, VEN, fork, enveloping, and other multipolar), VENs showed the greatest percentage that displayed immunostaining. Additionally, a higher proportion of VENs in humans were immunoreactive to ATF3, IL4Rα, and NMB than in other apes. No other ACC layer V neuron type displayed a significant species difference in the percentage of immunoreactive neurons. CONCLUSIONS: These findings demonstrate that phylogenetic variation exists in the protein expression profile of VENs, suggesting that humans might have evolved biochemical specializations for enhanced interoceptive sensitivity.


Subject(s)
Cerebral Cortex/physiology , Hominidae/physiology , Neurons/physiology , Activating Transcription Factor 3/physiology , Adult , Animals , Cell Count , Female , Hominidae/classification , Humans , Hylobatidae/physiology , Imaging, Three-Dimensional , Immunohistochemistry , Male , Middle Aged , Neurokinin B/analogs & derivatives , Neurokinin B/physiology , Neurons/classification , Receptors, Interleukin-4/physiology , Social Behavior , Young Adult
17.
Br J Ophthalmol ; 94(9): 1239-43, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20610477

ABSTRACT

AIM: To investigate the expression and function of interleukin-4 receptor alpha (IL-4Ralpha) in human conjunctival epithelial cells (HCjECs). METHODS: The presence of IL-4Ralpha mRNA and protein was examined by reverse transcription (RT) PCR and immunohistology, respectively. Cell surface expression was examined by flow cytometry. The effects of interleukin (IL)-4 or IL-13 on the tyrosine phosphorylation of signal transducer and the activator of transcription 6 (STAT6) were evaluated by immunoblot analyses. The transcripts upregulated upon IL-4 stimulation were examined using GeneChip, and confirmed by quantitative RT-PCR. RESULTS: IL-4Ralpha mRNA and protein were detected in human conjunctival epithelium. IL-4Ralpha protein was expressed on the cell surface of HCjECs. IL-4 and IL-13 induced tyrosine phosphorylation of STAT6. GeneChip analysis showed that nine transcripts were upregulated more than fourfold by IL-4 stimulation in the primary HCjECs from two individuals. Quantitative RT-PCR assay confirmed the upregulation of these transcripts: lecithin retinol acyltransferase (LRAT), calpain (CAPN14), tumour necrosis factor alpha-induced protein 6 (TNFAIP6), RAS guanyl-releasing protein 1 (RASGRP1), endothelin receptor type A (EDNRA), hyaluronan synthase 3 (HAS3), cathepsin C (CTSC), carbonic anhydrase II (CA2) and cytokine-inducible SH2-containing protein (CISH). CONCLUSIONS: HCjECs expressed functioning IL-4Ralpha, and IL-4 stimulation induced the expression of several genes.


Subject(s)
Conjunctiva/metabolism , Epithelial Cells/metabolism , Receptors, Interleukin-4/physiology , Calpain/metabolism , Conjunctiva/cytology , Humans , Immunohistochemistry , Interleukin-13/pharmacology , Interleukin-4/metabolism , Interleukin-4/pharmacology , Leukocytes, Mononuclear/metabolism , Phosphorylation , Receptors, Interleukin-4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT6 Transcription Factor/metabolism , Transcription Factors/metabolism , Up-Regulation
18.
J Ethnopharmacol ; 131(1): 104-9, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20561578

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: San-ao Decoction (SA) is a classical prescription, clinically employed to treat asthma in Chinese medicine. AIM OF STUDY: The present study was designed to examine whether SA has a protective effect on normal human bronchial epithelium modeled by interleukin-4 (IL-4), in association with eotaxin-3. MATERIALS AND METHODS: SA is made of three traditional Chinese medicines: Herba Ephedrae, Semen Armeniacae amarum and Radix Glycyrrhizae. Apoptosis was measured by fluorescence microscopy and flow cytometry with IL-4 activated NHBE. In addition, eotaxin-3 mRNA's expression was detected by RT-PCR in NHBE stimulated with IL-4. RESULTS: Fluorescence microscopy and flow cytometry showed that IL-4-induced normal human bronchial epithelium (NHBE) apoptosis, while SA decreased the apoptosis of NHBE with IL-4 stimulation. RT-PCR showed no expression or low expression of eotaxin-3 mRNA on NHBE, IL-4 enhanced the eotaxin-3 mRNA's expression, and that could be decreased by SA. CONCLUSIONS: The results indicate that SA can decrease NHBE's damage and inflammation through reducing eotaxin-3 mRNA expression.


Subject(s)
Bronchi/drug effects , Drugs, Chinese Herbal/pharmacology , Interleukin-4/pharmacology , Respiratory Mucosa/drug effects , Apoptosis/drug effects , Apoptosis/physiology , Bronchi/cytology , Cell Line , Drugs, Chinese Herbal/isolation & purification , Humans , Random Allocation , Receptors, Interleukin-4/agonists , Receptors, Interleukin-4/physiology , Respiratory Mucosa/cytology
19.
J Immunol ; 184(9): 4688-95, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20304823

ABSTRACT

Mast cell responses can be altered by cytokines, including those secreted by Th2 and regulatory T cells (Treg). Given the important role of mast cells in Th2-mediated inflammation and recent demonstrations of Treg-mast cell interactions, we examined the ability of IL-4 and TGF-beta1 to regulate mast cell homeostasis. Using in vitro and in vivo studies of mouse and human mast cells, we demonstrate that IL-4 suppresses TGF-beta1 receptor expression and signaling, and vice versa. In vitro studies demonstrated that IL-4 and TGF-beta1 had balancing effects on mast cell survival, migration, and FcepsilonRI expression, with each cytokine cancelling the effects of the other. However, in vivo analysis of peritoneal inflammation during Nippostrongylus brasiliensis infection in mice revealed a dominant suppressive function for TGF-beta1. These data support the existence of a cytokine network involving the Th2 cytokine IL-4 and the Treg cytokine TGF-beta1 that can regulate mast cell homeostasis. Dysregulation of this balance may impact allergic disease and be amenable to targeted therapy.


Subject(s)
Homeostasis/immunology , Interleukin-4/physiology , Mast Cells/immunology , Mast Cells/metabolism , Transforming Growth Factor beta1/physiology , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/physiology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/physiology , Receptors, Interleukin-4/antagonists & inhibitors , Receptors, Interleukin-4/biosynthesis , Receptors, Interleukin-4/physiology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/physiology , Tissue Culture Techniques , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/biosynthesis
20.
J Surg Res ; 162(2): 290-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19540524

ABSTRACT

BACKGROUND: The Interleukin-4 signal transducer and activator of transcription 6 (IL-4-STAT6) signaling pathway plays a pivotal role in regulation of gene transcription. We have previously identified a defective STAT6 activational phenotype in response to IL-4 in patients from our familial Inflammatory Bowel Disease registry. This has been termed Stat6(null) and Stat6(high) is the normal phenotype. The purpose of this study was to investigate the defect in Stat6 activation in Stat6(null) cells. METHODS: Stat6(null) and Stat6(high) Epstein Barr virus transformed cell lines were stimulated with 10 ng/mL of IL-4 for 0, 10, 30, or 60 min and cytoplasmic and nuclear proteins harvested. Western blot for STAT6, phosphorylated STAT6 (pSTAT6), Janus Kinase (Jak)1 and Jak3 was performed. Cells were also cultured for 48 h and interferon gamma (IFNgamma) measured in the supernatant. Additional cells were cultured with 20 ng/mL of IFNgamma for 90 min or 5 ug of antibody to IFNgamma for 48 h, and then stimulated with IL-4. RESULTS: There were no differences in cytoplasmic STAT6 in Stat6(null)versus Stat6(high) cells. In Stat6(high) cells, STAT6 was rapidly phosphorylated and translocated to the nucleus. In Stat6(null) cells there was minimal phosphorylation and translocation of pSTAT6 to the nucleus. Spontaneous secretion of IFNgamma by Stat6(null) cells was significantly higher than Stat6(null) cells. Addition of IFNgamma decreased pSTAT6 in Stat6(high) cells to Stat6(null) levels while blocking IFNgamma increased baseline pSTAT6 in Stat6(null) cells to levels similar to Stat6(high) cells. CONCLUSION: This suggests that the spontaneously produced IFNgamma in the Stat6(null) cell lines suppresses STAT6 function and creates the Stat6(null) phenotype.


Subject(s)
B-Lymphocytes/physiology , Herpesvirus 4, Human/physiology , Interleukin-4/metabolism , Lymphocytes/physiology , Receptors, Interleukin-4/physiology , STAT6 Transcription Factor/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/virology , Blotting, Western , Cell Line , Cell Line, Transformed , Humans , Interferon-gamma/pharmacology , Interleukin-4/pharmacology , Janus Kinase 1/metabolism , Janus Kinase 3/metabolism , Lymphocytes/drug effects , Phosphorylation , Registries , STAT6 Transcription Factor/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...