Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 8(6): 1262-74, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25783969

ABSTRACT

The epithelial lining of the airway tract and allergen-specific IgE are considered essential controllers of inflammatory responses to allergens. The human low affinity IgE receptor, CD23 (FcɛRII), is capable of transporting IgE or IgE-allergen complexes across the polarized human airway epithelial cell (AEC) monolayer in vitro. However, it remains unknown whether the CD23-dependent IgE transfer pathway in AECs initiates and facilitates allergic inflammation in vivo, and whether inhibition of this pathway attenuates allergic inflammation. To this end, we show that in wild-type (WT) mice, epithelial CD23 transcytosed both IgE and ovalbumin (OVA)-IgE complexes across the airway epithelial barrier, whereas neither type of transcytosis was observed in CD23 knockout (KO) mice. In chimeric mice, OVA sensitization and aerosol challenge of WT/WT (bone-marrow transfer from the WT to WT) or CD23KO/WT (CD23KO to WT) chimeric mice, which express CD23 on radioresistant airway structural cells (mainly epithelial cells) resulted in airway eosinophilia, including collagen deposition and a significant increase in goblet cells, and increased airway hyperreactivity. In contrast, the absence of CD23 expression on airway structural or epithelial cells, but not on hematopoietic cells, in WT/CD23KO (the WT to CD23KO) chimeric mice significantly reduced OVA-driven allergic airway inflammation. In addition, inhalation of the CD23-blocking B3B4 antibody in sensitized WT mice before or during airway challenge suppressed the salient features of asthma, including bronchial hyperreactivity. Taken together, these results identify a previously unproven mechanism in which epithelial CD23 plays a central role in the development of allergic inflammation. Further, our study suggests that functional inhibition of CD23 in the airway is a potential therapeutic approach to inhibit the development of asthma.


Subject(s)
Asthma/immunology , Hypersensitivity/immunology , Immunoglobulin E/metabolism , Receptors, IgE/immunology , Transcytosis/immunology , Animals , Asthma/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Hypersensitivity/metabolism , Immunoglobulin E/immunology , Immunohistochemistry , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, IgE/metabolism , Reverse Transcriptase Polymerase Chain Reaction
2.
Int Immunopharmacol ; 17(3): 568-75, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23994348

ABSTRACT

Neuroimmune semaphorin 4A (Sema4A) has been shown to play an important costimulatory role in T cell activation and regulation of Th1-mediated diseases such as multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE), and experimental autoimmune myocarditis (EAM). Sema4A has three functional receptors, Tim-2 expressed on CD4+ T cells, Th2 cells in particular, and Plexin B1 and D1 predominantly expressed on epithelial and endothelial cells, correspondingly. We recently showed that Sema4A has a complex expression pattern in lung tissue in a mouse model of asthma. We and others have shown that corresponding Plexin expression can be found on immune cells as well. Moreover, we demonstrated that Sema4A-deficient mice displayed significantly higher lung local and systemic allergic responses pointing to its critical regulatory role in the disease. To determine the utility of Sema4A as a novel immunotherapeutic, we introduced recombinant Sema4A protein to the allergen-sensitized WT and Sema4A(-/-) mice before allergen challenge. We observed significant reductions in the allergic inflammatory lung response in Sema4A-treated mice as judged by tissue inflammation including eosinophilia and mucus production. Furthermore, we demonstrated that in vivo administration of anti-Tim2 Ab led to a substantial upregulation of allergic inflammation in WT mouse lungs. These data highlight the potential to develop Sema4A as a new therapeutic for allergic airway disease.


Subject(s)
Asthma/immunology , Semaphorins/immunology , Allergens/immunology , Animals , Antibodies/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Count , Cytokines/blood , Female , Granulocytes/immunology , Male , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Recombinant Proteins/pharmacology , Semaphorins/pharmacology
3.
Mol Immunol ; 56(4): 480-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23911404

ABSTRACT

Neuroimmune semaphorin 4D (Sema4D) was found to be expressed and function in the nervous and immune systems. In the immune system, Sema4D is constitutively expressed on T cells and regulates T cell priming. In addition, it displays a stimulatory function on macrophages, DC, NK cells, and neutrophils. As all these cells are deeply involved in asthma pathology, we hypothesized that Sema4D plays a critical non-redundant regulatory role in allergic airway response. To test our hypothesis, we exposed Sema4D(-/-) and WT mice to OVA injections and challenges in the well-defined mouse model of OVA-induced experimental asthma. We observed a significant decrease in eosinophilic airway infiltration in allergen-treated Sema4D(-/-) mice relative to WT mice. This reduced allergic inflammatory response was associated with decreased BAL IL-5, IL-13, TGFß1, IL-6, and IL-17A levels. In addition, T cell proliferation in OVA323₋339-restimulated Sema4D(-/-) cell cultures was downregulated. We also found increased Treg numbers in spleens of Sema4D(-/-) mice. However, airway hyperreactivity (AHR) to methacholine challenges was not affected by Sema4D deficiency in either acute or chronic experimental disease setting. Surprisingly, lung DC number and activation were not affected by Sema4D deficiency. These data provide a new insight into Sema4D biology and define Sema4D as an important regulator of Th2-driven lung pathophysiology and as a potential target for a combinatory disease immunotherapy.


Subject(s)
Antigens, CD/immunology , Hypersensitivity/immunology , Lung/immunology , Pneumonia/immunology , Semaphorins/immunology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Asthma/genetics , Asthma/immunology , Asthma/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cell Proliferation , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Flow Cytometry , Humans , Hypersensitivity/genetics , Hypersensitivity/metabolism , Lung/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Pneumonia/genetics , Pneumonia/metabolism , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/metabolism , Semaphorins/genetics , Semaphorins/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
4.
Mucosal Immunol ; 5(4): 409-19, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22472774

ABSTRACT

To define the role of semaphorin 4A (Sema4A) in allergic response, we employed Sema4A⁻/⁻ and wild-type (WT) mice in the experimental model of ovalbumin (OVA)-induced allergic airway inflammation. We observed a selective increase in eosinophilic airway infiltration accompanied by bronchial epithelial cell hyperplasia in allergen-treated Sema4A⁻/⁻ mice relative to WT mice. This enhanced inflammatory response was associated with a selective increase in bronchoalveolar lavage (BAL) interleukin 13 (IL-13) content, augmented airway hyperreactivity, and lower regulatory T cell (Treg) numbers. In vivo allergen-primed Sema4A⁻/⁻ CD4+ T cells were more effective in transferring T helper type 2 (Th2) response to naive mice as compared with WT CD4+ T cells. T-cell proliferation and IL-13 productions in OVA323₋339-restimulated Sema4A⁻/⁻ cell cultures were upregulated. Generated bone marrow chimeras showed an equal importance of both lung-resident cell and inflammatory cell Sema4A expression in optimal disease regulation. These data provide a new insight into Sema4A biology and define Sema4A as an important regulator of Th2-driven lung pathophysiology.


Subject(s)
Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology , Semaphorins/genetics , Animals , Asthma/genetics , Asthma/immunology , Bone Marrow/immunology , Bone Marrow/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/chemistry , Ovalbumin/immunology , Semaphorins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
5.
Allergy ; 66(9): 1193-200, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21557750

ABSTRACT

BACKGROUND: Elevated levels of CD23, a natural regulator of IgE production, have been shown to decrease the signs of lung inflammation in mice. The aim of this study was to study the involvement of ADAM10, the primary CD23 sheddase, in experimental asthma. METHODS: ADAM10 was blocked either by using mice with a B-cell-specific deletion of the protease or pharmacologically by intranasal administration of selective ADAM10 inhibitors. Airway hypersensitivity (AHR) and bronchoaveolar lavage fluid (BALF) eosinophilia and select BALF cytokine/chemokine levels were then determined. RESULTS: Using an IgE and mast cell-dependent mouse model, B-cell-specific ADAM10(-/-) mice (C57B/6 background) exhibited decreased eosinophilia and AHR when compared with littermate (LM) controls. Treatment of C57B/6 mice with selective inhibitors of ADAM10 resulted in an even further decrease in BALF eosinophilia, as compared with the ADAM10(-/-) animals. Even in the Th2 selective strain, Balb/c, BALF eosinophilia was reduced from 60% to 23% respectively. In contrast, when an IgE/mast cell-independent model of lung inflammation was used, the B-cell ADAM10(-/-) animals and ADAM10 inhibitor treated animals had lung inflammation levels that were similar to the controls. CONCLUSIONS: These results thus show that ADAM10 is important in the progression of IgE-dependent lung inflammation. The use of the inhibitor further suggested that ADAM10 was important for maintaining Th2 levels in the lung. These results thus suggest that decreasing ADAM10 activity could be beneficial in controlling asthma and possibly other IgE-dependent diseases.


Subject(s)
ADAM Proteins/antagonists & inhibitors , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Asthma/therapy , Membrane Proteins/antagonists & inhibitors , ADAM Proteins/genetics , ADAM Proteins/physiology , ADAM10 Protein , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/physiology , Animals , Asthma/immunology , Asthma/pathology , Disease Models, Animal , Female , Gene Expression Regulation/immunology , Immunoglobulin E/immunology , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pneumonia/immunology , Pneumonia/pathology , Receptors, IgE/genetics
6.
Mucosal Immunol ; 3(3): 291-300, 2010 May.
Article in English | MEDLINE | ID: mdl-20404812

ABSTRACT

Severe respiratory syncytial virus (RSV)-induced bronchiolitis has been associated with a mixed "Th1" and "Th2" cytokine storm. We hypothesized that differentiation of "alternatively activated" macrophages (AA-M phi) would mediate the resolution of RSV-induced lung injury. RSV induced interleukin (IL)-4 and IL-13 by murine lung and peritoneal macrophages, IL-4R alpha/STAT6-dependent AA-M phi differentiation, and significantly enhanced inflammation in the lungs of IL-4R alpha(-/-) mice. Adoptive transfer of wildtype macrophages to IL-4R alpha(-/-) mice restored RSV-inducible AA-M phi phenotype and diminished lung pathology. RSV-infected Toll-like receptor (TLR)4(-/-) and interferon (IFN)-beta(-/-) macrophages and mice also failed to express AA-M phi markers, but exhibited sustained proinflammatory cytokine production (e.g., IL-12) in vitro and in vivo and epithelial damage in vivo. TLR4 signaling is required for peroxisome proliferator-activated receptor gamma expression, a DNA-binding protein that induces AA-M phi genes, whereas IFN-beta regulates IL-4, IL-13, IL-4R alpha, and IL-10 expression in response to RSV. RSV-infected cotton rats treated with a cyclooxygenase-2 inhibitor increased expression of lung AA-M phi. These data suggest new treatment strategies for RSV that promote AA-M phi differentiation.


Subject(s)
Interferon-beta/immunology , Lung Injury/immunology , Macrophage Activation/immunology , Macrophages, Alveolar/immunology , Macrophages, Peritoneal/immunology , Receptors, Cell Surface/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Toll-Like Receptor 4/immunology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , Cyclooxygenase 2 Inhibitors/pharmacology , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Lung Injury/metabolism , Lung Injury/virology , Macrophage Activation/drug effects , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/virology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/virology , Mice , Mice, Inbred BALB C , Mice, Knockout , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/virology , Rats , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/metabolism , Sigmodontinae , Signal Transduction/drug effects , Signal Transduction/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
7.
Cell Death Differ ; 10(2): 203-10, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12700648

ABSTRACT

Upon activation, naïve T helper cells can differentiate into two major distinct subsets, T helper 1 (Th1) and T helper 2 (Th2), as defined by their effector functions and cytokine secretion patterns. Cytokine milieu and costimulatory molecules have been shown to play an essential role in determining T helper differentiation. However, it is still unclear how the effects of signals of costimulatory molecules and cytokines are exerted during T helper differentiation. We show evidence suggesting that while cytokine signals initiate the differentiation program, the selective action of death effectors determines the end point balance of differentiating T helper subsets. We examined the expression of TNF-related apoptosis-inducing ligand (TRAIL) and CD95L in cloned and in vitro differentiated Th1 and Th2 cells. We found that activation-induced expression of TRAIL is exclusively observed in Th2 clones and primary T helper cells differentiated under the Th2 condition, while the expression of CD95L is mainly in Th1 cells. Furthermore, these two subsets exhibit distinct susceptibilities to TRAIL- and CD95L-mediated apoptosis. Th2 cells are more resistant to either TRAIL- or CD95L-induced apoptosis than Th1 cells. More importantly, both Th1 and Th2 cells could induce apoptosis in labeled Th1 but not Th2 cells. Blocking TRAIL and CD95L significantly enhance IFN-gamma production in vitro. Likewise, young MRL/MpJ-lpr/lpr mice also showed more Th1 response to ovalbumin immunization as compared to MRL/MpJ+/+. Therefore, apoptosis mediated by CD95L and TRAIL is critical in determining the fate of differentiating T helper cells.


Subject(s)
Apoptosis , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis Regulatory Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/metabolism , Cell Differentiation , Cells, Cultured , Clone Cells , Fas Ligand Protein , Female , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred Strains , Spleen/cytology , TNF-Related Apoptosis-Inducing Ligand , Th1 Cells/immunology , Th2 Cells/immunology
8.
Int Immunol ; 13(12): 1479-87, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11717189

ABSTRACT

IL-4 alone protects cells from apoptosis by insulin receptor substrate (IRS)-dependent and -independent mechanisms. However, in vivo cells are typically exposed to a number of signals at the same time. To determine the contribution of co-stimulatory signals to the regulation of apoptosis by IL-4, we first analyzed whether tumor necrosis factor (TNF)-alpha, which has been shown to inhibit the activation of IRS-1 by insulin, could modify IL-4 signaling and protection from apoptosis. We found that TNF-alpha cooperates with IL-4 in protecting 32D cells from factor withdrawal-induced apoptosis. This effect was independent of the expression of IRS-1, indicating that this cooperation is via an alternative anti-apoptotic pathway. Moreover, TNF-alpha had no effect on the activation of IRS-1 induced by IL-4. IL-4 enhanced TNF-alpha-induced activation of the transcription factor NF-kappaB. Interestingly, pharmacologic inhibition of NF-kappaB activation or protein synthesis resulted in the induction of cell death that could not be inhibited by IL-4, suggesting that IL-4 cooperates with NF-kappaB to signal protection from apoptosis. Supporting this hypothesis, IL-4 also increased NF-kappaB activation induced by anti-CD3 antibodies in primary T cells and protected them from apoptosis induced by receptor engagement. However, IL-4 was not able to inhibit apoptosis induced by anti-CD3 in T lymphocytes isolated from transgenic mice expressing a dominant-negative form of IkappaBalpha that prevents NF-kappaB activation. Thus, in addition to the previously identified IRS-1 pathway, IL-4-induced protection from apoptosis may also be mediated through cooperation with the NF-kappaB family of transcription factors.


Subject(s)
Apoptosis/immunology , Interleukin-4/physiology , NF-kappa B/metabolism , Adjuvants, Immunologic/physiology , Animals , Antibodies, Monoclonal/pharmacology , CD3 Complex/immunology , Cell Survival/immunology , Insulin Receptor Substrate Proteins , Mice , Mice, Transgenic , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Phosphoproteins/metabolism , Receptor, Insulin/metabolism , STAT6 Transcription Factor , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Trans-Activators/metabolism , Tumor Necrosis Factor-alpha/physiology
9.
Gene Ther ; 8(12): 938-45, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11426334

ABSTRACT

Murine lymphocytes are relatively refractory to efficient transfection or retroviral gene transduction. Adenovirus has been used as a vector to transduce a wide variety of cell types. Several advantages of adenoviruses are their ability to transduce non-cycling cells and to transduce the majority of cells in a population. Unfortunately, lymphocytes are not susceptible to infection with conventional adenovirus. Therefore, to express genes efficiently in murine B cells, we tested the ability of genetically modified adenovirus to transduce the beta-galactosidase gene. We found that adenovirus containing polylysine in the fiber knob was able to efficiently transduce lipopolysaccharide (LPS)-activated splenic B cells and the B lymphoma line M12.4.1; greater than 80% of the cells expressed beta-galactosidase activity. However, small resting B cells did not express activity unless treated with LPS after infection. This transduction was mediated by interaction with charged molecules since heparan-sulfate, and to a lesser degree chondroitan sulfate, inhibited the transduction. In addition, adenovirus containing a FLAG epitope in the fiber protein was used to target the FcR expressed on B cells using an anti-FLAG antibody. In the presence of anti-FLAG, the modified adenovirus was able to efficiently transduce LPS-activated B cells and several B cell lymphoma lines. Interestingly, in the absence of anti-FLAG, there was low level transduction in the LPS-blasts and in M12.4.1 that was not inhibited by soluble adenovirus fiber protein or agents that block RGD-integrin interactions. These results demonstrate that modified adenovirus efficiently transduce B lymphocytes which will be critical for targeting genes to normal or malignant B cells.


Subject(s)
Adenoviridae/genetics , B-Lymphocytes/metabolism , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Transduction, Genetic/methods , Animals , Gene Expression , HeLa Cells , Humans , Lipopolysaccharides/pharmacology , Lymphoma, B-Cell/therapy , Mice , Oligopeptides , Peptides/genetics , Tumor Cells, Cultured , beta-Galactosidase/genetics
10.
Cell Res ; 11(1): 44-54, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11305324

ABSTRACT

IL-4 is an important B cell survival and growth factor. IL-4 induced the tyrosine phosphorylation of IRS2 in resting B lymphocytes and in LPS- or CD40L-activated blasts. Phosphorylated IRS2 coprecipitated with the p85 subunit of PI 3' kinase in both resting and activated cells. By contrast, association of phosphorylated IRS2 with GRB2 was not detected in resting B cells after IL-4 treatment although both proteins were expressed. However, IL-4 induced association of IRS2 with GRB2 in B cell blasts. The pattern of IL-4-induced recruitment of p85 and GRB2 to IRS2 observed in B cells derived from STAT6 null mice was identical to that observed for normal mice. While IL-4 alone does not induce activation of MEK, a MEK1 inhibitor suppressed the IL-4-induced proliferative response of LPS-activated B cell blasts. These results demonstrate that costimulation of splenic B cells alters IL-4-induced signal transduction independent of STAT6 leading to proliferation. Furthermore, proliferation induced by IL-4 in LPS-activated blasts is dependent upon the MAP kinase pathway.


Subject(s)
Adaptor Proteins, Signal Transducing , B-Lymphocytes/metabolism , Cell Division/physiology , Cell Survival/physiology , Interleukin-4/metabolism , Phosphoproteins/metabolism , Signal Transduction/physiology , Trans-Activators/deficiency , Animals , Apoptosis/drug effects , Apoptosis/physiology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CD40 Ligand/pharmacology , Cell Division/drug effects , Cell Survival/drug effects , Cells, Cultured/drug effects , Cells, Cultured/immunology , Cells, Cultured/metabolism , Enzyme Inhibitors/pharmacology , GRB2 Adaptor Protein , Insulin Receptor Substrate Proteins , Interleukin-4/pharmacology , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/pharmacology , MAP Kinase Kinase 1 , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/drug effects , Phosphoproteins/isolation & purification , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Proteins/drug effects , Proteins/metabolism , STAT6 Transcription Factor , Signal Transduction/drug effects , Trans-Activators/genetics
11.
Annu Rev Immunol ; 17: 701-38, 1999.
Article in English | MEDLINE | ID: mdl-10358772

ABSTRACT

Interleukin-4 is a multifunctional cytokine that plays a critical role in the regulation of immune responses. Its effects depend upon binding to and signaling through a receptor complex consisting of the IL-4R alpha chain and the common gamma chain (gamma c), resulting in a series of phosphorylation events mediated by receptor-associated kinases. In turn, these cause the recruitment of mediators of cell growth, of resistance to apoptosis, and of gene activation and differentiation. Here we describe our current understanding of the organization of the IL-4 receptor, of the signaling pathways that are induced as a result of receptor occupancy, and of the various mechanisms through which receptor function is modulated. We particularly emphasize the modular nature of the receptor and the specialization of different receptor regions for distinct functions, most notably the independent regulation of cell growth and gene activation.


Subject(s)
Mitogen-Activated Protein Kinases , Receptors, Interleukin-4/physiology , Amino Acid Sequence , Animals , Calcium-Calmodulin-Dependent Protein Kinases/immunology , Humans , Insulin Receptor Substrate Proteins , Interleukin-4/physiology , Intracellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases , Mice , Mice, Knockout , Molecular Sequence Data , Phosphoproteins/immunology , Protein Tyrosine Phosphatases/immunology , Receptors, Interleukin-4/chemistry , Receptors, Interleukin-4/genetics , STAT6 Transcription Factor , Signal Transduction , Trans-Activators/genetics , Trans-Activators/immunology
12.
J Immunol ; 162(8): 4385-9, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10201973

ABSTRACT

A mutation in the human (hu) IL-4R alpha, Q576R, has been linked with allergy in humans. Increased sensitivity of patients cells with this mutation to IL-4 suggest that a Q576R change enhances IL-4 signaling. To directly test this hypothesis, we analyzed the ability of huIL-4R alpha cDNA bearing the Q576R and Y575F mutations to signal tyrosine phosphorylation, DNA-binding activity, proliferation, protection from apoptosis, and CD23 induction in response to huIL-4 in murine cells. Responses generated by the Q576R and Y575F mutants were similar to those of the wild-type receptor, using various concentrations of huIL-4 and times of stimulation. These results indicate that neither the Q576R nor the Y575F mutations have a significant direct effect on IL-4 signal transduction, and that hypersensitive induction of CD23 in cells derived from human allergy patients may be due to different and/or additional alterations in the IL-4 signaling pathway.


Subject(s)
Hypersensitivity/genetics , Mutagenesis, Site-Directed/immunology , Receptors, Interleukin-4/genetics , Signal Transduction/immunology , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Arginine/genetics , Cell Line , DNA-Binding Proteins/biosynthesis , Glutamine/genetics , Humans , Hypersensitivity/immunology , Insulin Receptor Substrate Proteins , Lymphocyte Activation/genetics , Mice , Phosphoproteins/metabolism , Phosphorylation , Receptor, Insulin/metabolism , Receptors, IgE/biosynthesis , Receptors, Interleukin-4/physiology , STAT6 Transcription Factor , Signal Transduction/genetics , Trans-Activators/metabolism , Tyrosine/genetics , Tyrosine/metabolism
13.
J Immunol ; 161(2): 859-67, 1998 Jul 15.
Article in English | MEDLINE | ID: mdl-9670964

ABSTRACT

IL-4 is a cytokine with important antiapoptotic activity. We have analyzed the role that tyrosine-containing domains within the cytoplasmic tail of IL-4R alpha play in IL-4-mediated protection from apoptosis. 32D cells expressing a wt huIL-4R alpha or one truncated at aa 557 were protected by huIL-4 from apoptosis while cells expressing a receptor truncated at aa 657 were not, suggesting that the carboxyl-terminal domain signals protection from apoptosis. However, changing Y713 within this region to phenylalanine had no effect. To analyze the contribution of tyrosine-containing domains independently, we transplanted regions of the huIL-4R alpha to a truncated form of the huIL-2R beta that could not signal protection from apoptosis. Transplantation of the huIL-4R alpha domains containing Y497 or Y713 partially prevented cell death and together signaled protection from apoptosis in response to IL-2 as well as the wt IL-2R beta. Mutation of Y497 and Y713 to phenylalanine inhibited protection. In contrast, transplantation of the domain containing the potential STAT6-docking tyrosines alone had no effect, yet it inhibited the protection mediated by the other domains. Although IL-4R alpha signals Shc and SH2-containing inositol phosphatase (SHIP) phosphorylation, we could not establish an association between their activation and protection from apoptosis. Taken together, this study suggests that the domains of the huIL-4R alpha containing Y497 and Y713 positively regulate protection from apoptosis while the domain containing the STAT6 docking sites suppresses this protection, and that additional signaling molecules other than insulin receptor substrate-1 (IRS1), Shc, or SHIP may be involved in antiapoptotic signaling.


Subject(s)
Apoptosis/immunology , Receptors, Interleukin-4/physiology , Signal Transduction/immunology , Trans-Activators/physiology , Tyrosine/physiology , Animals , Apoptosis/genetics , Humans , Insulin Receptor Substrate Proteins , Mice , Mutagenesis, Site-Directed/immunology , Peptide Fragments/genetics , Peptide Fragments/physiology , Phenylalanine/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Structure, Tertiary , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/physiology , Receptors, Interleukin-4/genetics , STAT6 Transcription Factor , Signal Transduction/genetics , Tumor Cells, Cultured , Tyrosine/genetics , src Homology Domains/immunology
14.
J Biol Chem ; 273(16): 9898-905, 1998 Apr 17.
Article in English | MEDLINE | ID: mdl-9545332

ABSTRACT

The interleukin (IL)-4 receptor alpha-chain (IL-4Ralpha) contains a sequence motif (488PLVIAGNPAYRSFSD) termed the insulin IL-4 receptor motif (I4R motif). Mutation of the central Tyr497 to Phe blocks the tyrosine phosphorylation of the insulin receptor substrate 1 (IRS1) and diminishes proliferation in response to IL-4. Recent data suggest that the I4R motif encodes binding sites for several protein tyrosine binding (PTB) domain-containing proteins such as IRS1 and Shc and potentially for the Src homology 2 domain of signal transducer and activator of transcription 6 (STAT6). To analyze the function of the I4R motif in regulating IL-4 signaling, we changed conserved residues upstream and downstream of the central Tyr to Ala in the human IL-4Ralpha. We analyzed the ability of these constructs to signal the tyrosine phosphorylation of IRS2 and STAT6, the induction of DNA binding activity, and CD23 induction in response to human IL-4 (huIL-4) in transfected M12.4.1 cells. Mutagenesis of residues downstream of Tyr497, such as Arg498 or Phe500, to Ala had no effect on any of these responses, suggesting that the I4R motif may not be important for functional Src homology 2 domain interactions. However, mutagenesis of Pro488 to Ala (P488A) greatly diminished the tyrosine phosphorylation of IRS2 and abolished tyrosine phosphorylation of STAT6, induction of DNA binding activity, and CD23 induction in response to huIL-4. By contrast, a P488G mutant signaled these responses to huIL-4. Mutagenesis of hydrophobic amino acids previously shown to contact the PTB domain of IRS1, Leu489 or Ile491, to Ala had only minimal effects on responses to huIL-4. However, changing both Leu498 and Ile491 to Ala greatly diminished the tyrosine phosphorylation of IRS2 and abolished STAT6 activation. Taken together, these results indicate the important role of the I4R motif in regulating IRS docking and suggest that I4R docking to a PTB domain-containing protein regulates activation of the STAT6 pathway.


Subject(s)
DNA-Binding Proteins/metabolism , Interleukin-4/pharmacology , Phosphoproteins/metabolism , Receptors, Interleukin-4/physiology , Signal Transduction , Trans-Activators/metabolism , Alanine , Amino Acid Sequence , Animals , Conserved Sequence , DNA-Binding Proteins/biosynthesis , Humans , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Lymphoma, B-Cell , Macromolecular Substances , Mice , Models, Biological , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphotyrosine/metabolism , Point Mutation , Receptor, Insulin/physiology , Receptors, IgE/biosynthesis , Receptors, Interleukin-4/chemistry , Recombinant Proteins/pharmacology , STAT4 Transcription Factor , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Tumor Cells, Cultured , Tyrosine , src Homology Domains
15.
Cell Res ; 8(1): 1-13, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9570012

ABSTRACT

Interleukin-4 is a cytokine produced by activated T cells, mast cells, and basophils that elicits many important biological responses[1] (see Tab 1). These responses range from the regulation of helper T cell differentiation[2] and the production of IgE[3] to the regulation of the adhesive properties of endothelial cells via VCAM-1[4]. In keeping with these diverse biological effects, high-affinity binding sites for IL-4 (Kd 20 to 300 pM) have been detected on many hematopoietic and non-hematopoietic cell types at levels ranging from 50 to 5000 sites per cell[5]. This review will focus on the discrete signal transduction pathways activated by the IL-4 receptor and the coordination of these individual pathways in the regulation of a final biological outcome.


Subject(s)
Interleukin-4/physiology , Receptors, Interleukin-4/physiology , Signal Transduction/physiology , Amino Acid Sequence , Cell Division , Gene Expression Regulation/physiology , Humans , Lymphocytes/cytology , Molecular Sequence Data
16.
J Immunol ; 160(7): 3502-12, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9531312

ABSTRACT

Cytokines play an essential role in the regulation of lymphocyte survival and growth. We have analyzed the pathways activated by IE-2 that lead to protection from apoptosis and cell proliferation. IL-2 can act as a long-term growth factor in 32D cells expressing the wild-type human (hu)IL-2R beta. By contrast, cells expressing a truncated form of the huIL-2R beta, which is able to induce Bcl-2 and c-myc expression but not STAT5 activation, were not protected from apoptosis by IL-2; consequently, they could not be grown long term in the presence of IL-2. However, IL-2 promoted cell cycle progression in cells bearing the truncated huIL-2R beta with percentages of viable cells in the G0/G1, S, and G2/M phases similar to cells expressing the wild-type huIL-2R beta. Transplantation of a region from the erythropoietin receptor, which contains a docking site for STAT5 (Y343) to the truncated huIL-2R beta, restored the ability of IL-2 to signal both activation of STAT5 and protection from apoptosis. By contrast, transplantation of a region from the huIL-4R alpha containing STAT6 docking sites did not confer protection from apoptosis. These results indicate that the IL-2-induced cell cycle progression can be clearly distinguished from protection from apoptosis and that STAT5 participates in the regulation of apoptosis.


Subject(s)
Apoptosis/immunology , Cell Cycle/immunology , Interleukin-2/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Cell Division/genetics , Cell Division/immunology , Humans , Mice , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Cells, Cultured
17.
J Biol Chem ; 272(40): 25083-90, 1997 Oct 03.
Article in English | MEDLINE | ID: mdl-9312117

ABSTRACT

Interleukin (IL)-4 is a cytokine that regulates both the growth and differentiation of hematopoietic cells. Its ligand binding specificity and important signal transduction mechanisms are conferred by the IL-4 receptor alpha chain (IL-4Ralpha). The I4R is a tyrosine-containing motif within IL-4Ralpha that is critical for proliferative responses to IL-4. Although the I4R also contributes to gene regulation, nuclear targets directly regulated by this motif have not been described. It is shown here that the tyrosine at position 497 in the I4R is critical for regulation of the phosphorylation status of a set of nuclear proteins that includes HMG-I(Y), small non-histone chromosomal proteins involved in the control of gene expression in hematopoietic cell lines. Moreover, IL-4 is unable to induce HMG-I(Y) phosphorylation in insulin receptor substrate-1-deficient cells, and the inhibitor wortmannin completely blocks IL-4 regulation of HMG-I(Y) phosphorylation status but not activation of an IL-4 Stat protein. Taken together, these data indicate that HMG-I(Y) is a nuclear target whose phosphorylation status is regulated through the I4R motif via insulin receptor substrate proteins, independent of activation of the Stat pathway.


Subject(s)
Antigens, CD/metabolism , Cell Nucleus/metabolism , Hematopoietic Stem Cells/physiology , High Mobility Group Proteins/metabolism , Phosphoproteins/metabolism , Receptors, Interleukin/metabolism , Androstadienes/pharmacology , Animals , Antigens, CD/biosynthesis , Antigens, CD/chemistry , Cells, Cultured , Enzyme Induction , Enzyme Inhibitors/pharmacology , HMGA1a Protein , High Mobility Group Proteins/biosynthesis , Humans , Insulin Receptor Substrate Proteins , Macromolecular Substances , Mutagenesis, Site-Directed , Phosphoproteins/biosynthesis , Phosphorylation , Point Mutation , Rats , Receptors, Interleukin/biosynthesis , Receptors, Interleukin/chemistry , Receptors, Interleukin-4 , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , STAT6 Transcription Factor , Signal Transduction , Trans-Activators/biosynthesis , Transfection , Tyrosine , Wortmannin
18.
J Immunol ; 158(3): 1037-40, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9013940

ABSTRACT

It has been shown that IL-4 induces the tyrosine phosphorylation of JAK1 and JAK3 in the majority of hemopoietic cell types, and JAK2 and TYK2 in several other types. However, the significance of this tyrosine phosphorylation in regulating IL-4 signaling has not been shown. To determine whether JAKs play a role in activating a signal transduction pathway different from the classical JAK/STAT pathway, we analyzed the ability of huIL-4 to stimulate the tyrosine phosphorylation of one of its major cellular substrates, the insulin receptor substrate (IRS). Using human fibrosarcoma cell lines with mutations in JAK1, JAK2, and TYK2, we found that expression of functional JAK1, but not TYK2 or JAK2, is essential for IL-4-induced tyrosine phosphorylation of IRS. We also provide evidence that the IRS pathway is independent of STAT-6, showing that JAK1 is essential for activating a STAT-independent pathway.


Subject(s)
Interleukin-4/physiology , Phosphoproteins/metabolism , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/physiology , Fibrosarcoma/metabolism , Humans , Insulin Receptor Substrate Proteins , Janus Kinase 1 , Phosphorylation , STAT6 Transcription Factor , Signal Transduction , Trans-Activators/physiology , Tumor Cells, Cultured
19.
J Immunol ; 157(11): 4926-34, 1996 Dec 01.
Article in English | MEDLINE | ID: mdl-8943397

ABSTRACT

Although it has been known for some time that IL-4 protects cells from death via apoptosis, very little is known about the mechanism by which IL-4 exerts this effect. In this report, we have studied the molecular mechanisms of the IL-4-induced prevention of apoptosis in the 32D and B cell systems. IL-3 withdrawal has been shown to induce G1-arrest in 32D cells and subsequent death by apoptosis. We found that overexpression of IRS-1 protected 32D cells from death induced by IL-3 deprivation. IL-4 was able to protect 32D cells from apoptosis in the presence or absence of IRS-1. However, the protection from apoptosis of cells cultured in IL-4 was greater in the presence of IRS-1 and it reached levels comparable to those of cells cultured in IL-3. The IRS-1-dependent prevention of apoptosis is linked to the activation of PI 3'-kinase since wortmannin and LY294002, two inhibitors of PI 3'-K, partially inhibited the prevention of apoptosis mediated by IL-4 in 32D-IRS-1 cells after IL-3 withdrawal but not in 32D cells lacking IRS-1 expression. In addition, we found that the IRS/PI 3'-K pathway is, at least in part, responsible for the prevention of apoptosis by IL-4 in normal splenic B cell cultures. In spite of the ability of murine IL-4 to partially protect 32D cells lacking IRS-1 from apoptosis, human IL-4 was not able to prevent cell death in 32D-IRS-1 cells transfected with the human IL-4 receptor mutated in the insulin-lL-4 receptor motif (14R-motif) at amino acid 497 (Y497F). This mutation has previously been shown to abrogate the tyrosine phosphorylation of IRS-1 by human IL-4. These results demonstrate that IL-4 protects 32D cells from apoptosis by two different pathways, one of them mediated by IRS-1. In addition, these results suggest that the 14R-motif of the IL-4R is linked to both pathways.


Subject(s)
Apoptosis/drug effects , Interleukin-4/pharmacology , Phosphoproteins/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis/physiology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Base Sequence , Cell Line , DNA Primers/genetics , Humans , In Vitro Techniques , Insulin Receptor Substrate Proteins , Interleukin-4/genetics , Mice , Mutation , Phosphatidylinositol 3-Kinases , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Receptors, Interleukin-4 , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Ribosomal Protein S6 Kinases , Signal Transduction/drug effects , Signal Transduction/physiology , Transfection
20.
Immunity ; 4(2): 113-21, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8624802

ABSTRACT

IL-4 binds to a cell surface receptor complex that consists of the IL-4 binding protein (IL-4R alpha) and the gamma chain of the IL-2 receptor complex (gamma c). The receptors for IL-4 and IL-2 have several features in common; both use the gamma c as a receptor component, and both activate the Janus kinases JAK-1 and JAK-3. In spite of these similarities, IL-4 evokes specific responses, including the tyrosine phosphorylation of 4PS/IRS-2 and the induction of CD23. To determine whether sequences within the cytoplasmic domain of the IL-4R alpha specify these IL-4-specific responses, we transplanted the insulin IL-4 receptor motif (I4R motif) of the huIL-4R alpha to the cytoplasmic domain of a truncated IL-2R beta. In addition, we transplanted a region that contains peptide sequences shown to block Stat6 binding to DNA. We analyzed the ability of cells expressing these IL-2R-IL-4R chimeric constructs to respond to IL-2. We found that IL-4 function could be transplanted to the IL-2 receptor by these regions and that proliferative and differentiative functions can be induced by different receptor sequences.


Subject(s)
Antigens, CD/physiology , Interleukin-4/physiology , Receptors, Interleukin-2/physiology , Receptors, Interleukin/physiology , Amino Acid Sequence , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Interleukin-4/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphoma, B-Cell , Mice , Molecular Sequence Data , Rats , Receptors, Interleukin/chemistry , Receptors, Interleukin/genetics , Receptors, Interleukin-2/genetics , Receptors, Interleukin-4 , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...