Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Biochem J ; 463(3): 393-403, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25137390

ABSTRACT

Interleukin-3 (IL-3) is a cytokine secreted by mast cells and activated T-cells known to be an important regulator of differentiation, survival, proliferation and activation of a range of haemopoietic lineages. The effects of IL-3 on target cells are mediated by a transmembrane receptor system composed of a cytokine-specific α-subunit and a ß-subunit, the principal signalling entity. In the mouse, two ß-subunits have co-evolved: a common ß-subunit (ßc) shared between IL-3 and the related cytokines IL-5 and granulocyte/macrophage colony-stimulating factor (GM-CSF); and an IL-3-specific ß-subunit (ßIL-3). ßIL-3 differs from ßc in its specificity for IL-3 and its capacity to bind IL-3 directly in the absence of an α-subunit, and, in the absence of structural information, the basis for these properties has remained enigmatic. In the present study, we have solved the crystal structure of the ßIL-3 ectodomain at 3.45 Å (1 Å=0.1 nm) resolution. This structure provides the first evidence that ßIL-3 adopts an arch-shaped intertwined homodimer with similar topology to the paralogous ßc structure. In contrast with apo-ßc, however, the ligand-binding interface of ßIL-3 appears to pre-exist in a conformation receptive to IL-3 engagement. Molecular modelling of the IL-3-ßIL-3 interface, in conjunction with previous mutational studies, suggests that divergent evolution of both ßIL-3 and IL-3 underlies their unique capacity for direct interaction and specificity.


Subject(s)
Cytokine Receptor Common beta Subunit/chemistry , Interleukin-3/chemistry , Animals , Crystallography, X-Ray , Cytokine Receptor Common beta Subunit/metabolism , Evolution, Molecular , Interleukin-3/metabolism , Mice , Models, Molecular , Protein Conformation , Protein Multimerization , Protein Structure, Tertiary , Recombinant Proteins/chemistry
2.
PLoS One ; 9(5): e98139, 2014.
Article in English | MEDLINE | ID: mdl-24848502

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) play important roles in leukocyte differentiation, although those utilised for specific programs and key functions remain incompletely characterised. As a global approach to gain insights into the potential regulatory role of miRNA in mast cell differentiation we characterised expression in BM cultures from the initiation of differentiation. In cultures enriched in differentiating mast cells we characterised miRNA expression and identified miRNA targeting the mRNA of putative factors involved in differentiation pathways and cellular identity. Detailed pathway analysis identified a unique miRNA network that is intimately linked to the mast cell differentiation program. METHODOLOGY/PRINCIPAL FINDINGS: We identified 86 unique miRNAs with expression patterns that were up- or down- regulated at 5-fold or more during bone marrow derived mast cells (BMMC) development. By employing TargetScan and MeSH databases, we identified 524 transcripts involved in 30 canonical pathways as potentially regulated by these specific 86 miRNAs. Furthermore, by applying miRanda and IPA analyses, we predict that 7 specific miRNAs of this group are directly associated with the expression of c-Kit and FcεRIα and likewise, that 18 miRNAs promote expression of Mitf, GATA1 and c/EBPα three core transcription factors that direct mast cell differentiation. Furthermore, we have identified 11 miRNAs that may regulate the expression of STATs-3, -5a/b, GATA2 and GATA3 during differentiation, along with 13 miRNAs that target transcripts encoding Ndst2, mMCP4 and mMCP6 and thus may regulate biosynthesis of mast cell secretory mediators. CONCLUSIONS/SIGNIFICANCE: This investigation characterises changes in miRNA expression in whole BM cultures during the differentiation of mast cells and predicts functional links between miRNAs and their target mRNAs for the regulation of development. This information provides an important resource for further investigations of the contributions of miRNAs to mast cell differentiation and function.


Subject(s)
Bone Marrow Cells/cytology , Gene Expression Profiling , Gene Expression Regulation , Mast Cells/cytology , MicroRNAs/metabolism , Animals , Cell Differentiation/genetics , Cytokines/metabolism , Femur/metabolism , Gene Regulatory Networks , Interleukin-3 Receptor alpha Subunit/metabolism , Leukocytes/cytology , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-kit/metabolism , Receptors, IgE/metabolism , STAT Transcription Factors/metabolism
3.
PLoS One ; 9(5): e97537, 2014.
Article in English | MEDLINE | ID: mdl-24824797

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are small non-coding RNAs that regulate complex transcriptional networks underpin immune responses. However, little is known about the specific miRNA networks that control differentiation of specific leukocyte subsets. In this study, we profiled miRNA expression during differentiation of eosinophils from bone marrow (BM) progenitors (bmEos), and correlated expression with potential mRNA targets involved in crucial regulatory functions. Profiling was performed on whole BM cultures to document the dynamic changes in miRNA expression in the BM microenvironment over the differentiation period. miRNA for network analysis were identified in BM cultures enriched in differentiating eosinophils, and chosen for their potential ability to target mRNA of factors that are known to play critical roles in eosinophil differentiation pathways or cell identify. METHODOLOGY/PRINCIPAL FINDINGS: We identified 68 miRNAs with expression patterns that were up- or down- regulated 5-fold or more during bmEos differentiation. By employing TargetScan and MeSH databases, we identified 348 transcripts involved in 30 canonical pathways as potentially regulated by these miRNAs. Furthermore, by applying miRanda and Ingenuity Pathways Analysis (IPA), we identified 13 specific miRNAs that are temporally associated with the expression of IL-5Rα and CCR3 and 14 miRNAs associated with the transcription factors GATA-1/2, PU.1 and C/EBPε. We have also identified 17 miRNAs that may regulate the expression of TLRs 4 and 13 during eosinophil differentiation, although we could identify no miRNAs targeting the prominent secretory effector, eosinophil major basic protein. CONCLUSIONS/SIGNIFICANCE: This is the first study to map changes in miRNA expression in whole BM cultures during the differentiation of eosinophils, and to predict functional links between miRNAs and their target mRNAs for the regulation of eosinophilopoiesis. Our findings provide an important resource that will promote the platform for further understanding of the role of these non-coding RNAs in the regulation of eosinophil differentiation and function.


Subject(s)
Bone Marrow Cells/physiology , Cell Differentiation/physiology , Eosinophils/metabolism , Eosinophils/physiology , Gene Expression Regulation/genetics , MicroRNAs/metabolism , RNA, Messenger/metabolism , Analysis of Variance , Animals , Azure Stains , Bone Marrow Cells/metabolism , DNA Primers/genetics , Flow Cytometry , Gene Expression Profiling , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Microarray Analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Specific Pathogen-Free Organisms
4.
PLoS One ; 8(8): e74376, 2013.
Article in English | MEDLINE | ID: mdl-23991218

ABSTRACT

Human interleukin-3 (hIL-3) is a polypeptide growth factor that regulates the proliferation, differentiation, survival and function of hematopoietic progenitors and many mature blood cell lineages. Although recombinant hIL-3 is a widely used laboratory reagent in hematology, standard methods for its preparation, including those employed by commercial suppliers, remain arduous owing to a reliance on refolding insoluble protein expressed in E. coli. In addition, wild-type hIL-3 is a poor substrate for radio-iodination, which has been a long-standing hindrance to its use in receptor binding assays. To overcome these problems, we developed a method for expression of hIL-3 in E. coli as a soluble protein, with typical yields of >3mg of purified hIL-3 per litre of shaking microbial culture. Additionally, we introduced a non-native tyrosine residue into our hIL-3 analog, which allowed radio-iodination to high specific activities for receptor binding studies whilst not compromising bioactivity. The method presented herein provides a cost-effective and convenient route to milligram quantities of a hIL-3 analog with wild-type bioactivity that, unlike wild-type hIL­3, can be efficiently radio-iodinated for receptor binding studies.


Subject(s)
Escherichia coli/genetics , Interleukin-3/biosynthesis , Amino Acid Sequence , Animals , Base Sequence , Chromatography, Reverse-Phase , DNA Primers , Humans , Interleukin-3/chemistry , Interleukin-3/genetics , Interleukin-3/physiology , Mass Spectrometry , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid , Solubility
5.
Biochemistry ; 50(13): 2464-77, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21329364

ABSTRACT

Interleukin-3 (IL-3), a cytokine produced primarily by activated T-cells during immune responses, is a crucial regulator of allergic inflammation. The three-dimensional structure of murine IL-3 (mIL-3) has remained elusive owing to its poor solubility and strong tendency toward aggregation under solution conditions typically used for structural studies. Here we describe the solution properties and structure of mIL-3 determined by NMR using an engineered construct of mIL-3 (mIL-3(33-156)). mIL-3 adopts a four-helical bundle fold, typical of proteins belonging to the short-chain cytokine family, and features a core of highly conserved hydrophobic residues. While significant line broadening and peak disappearance were observed in NMR spectra at higher temperatures, there was no evidence for temperature-dependent changes of the oligomeric state of mIL-3(33-156). Further analysis of the temperature dependence of amide (1)H chemical shifts and backbone (15)N relaxation parameters, including (15)N relaxation dispersion, revealed the presence of significant conformational exchange and local conformational heterogeneity. Residues recently shown by mutagenesis to play key roles in ß(IL-3) receptor recognition and activation, which are located within the α(A) and α(C) helices and aligned on one face of the mIL-3(33-156) structure, are relatively rigid. In contrast, pronounced conformational heterogeneity was observed for a cluster of residues located on the opposite side of mIL-3, which corresponds spatially to sites in the related cytokines human IL-3, IL-5, and GM-CSF that are known to mediate interactions with their respective α-receptor subunits. Such conformational heterogeneity may facilitate the interaction of mIL-3 with each of two naturally occurring mIL-3Rα isoforms, leading to structurally distinct high-affinity complexes.


Subject(s)
Interleukin-3/chemistry , Peptide Fragments/chemistry , Animals , Hydrogen-Ion Concentration , Interleukin-3/genetics , Mice , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/genetics , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Solubility , Temperature
6.
J Biol Chem ; 285(32): 24759-68, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20516062

ABSTRACT

The cytokines, interleukin-3 (IL-3), interleukin-5 (IL-5), and granulocyte-macrophage colony-stimulating factor (GM-CSF), exhibit overlapping activities in the regulation of hematopoietic cells. In humans, the common beta (betac) receptor is shared by the three cytokines and functions together with cytokine-specific alpha subunits in signaling. A widely accepted hypothesis is that receptor activation requires heterodisulfide formation between the domain 1 D-E loop disulfide in human betac (hbetac) and unidentified cysteine residues in the N-terminal domains of the alpha receptors. Since the development of this hypothesis, new data have been obtained showing that domain 1 of hbetac is part of the cytokine binding epitope of this receptor and that an IL-3Ralpha isoform lacking the N-terminal Ig-like domain (the "SP2" isoform) is competent for signaling. We therefore investigated whether distortion of the domain 1-domain 4 ligand-binding epitope in hbetac and the related mouse receptor, beta(IL-3), could account for the loss of receptor signaling when the domain 1 D-E loop disulfide is disrupted. Indeed, mutation of the disulfide in hbetac led to both a complete loss of high affinity binding with the human IL-3Ralpha SP2 isoform and of downstream signaling. Mutation of the orthologous residues in the mouse IL-3-specific receptor, beta(IL-3), not only precluded direct binding of mouse IL-3 but also resulted in complete loss of high affinity binding and signaling with the mouse IL-3Ralpha SP2 isoform. Our data are most consistent with a role for the domain 1 D-E loop disulfide of hbetac and beta(IL-3) in maintaining the precise positions of ligand-binding residues necessary for normal high affinity binding and signaling.


Subject(s)
Interleukin-3/chemistry , Receptors, Interleukin-3/physiology , Amino Acid Sequence , Animals , COS Cells , Cell Proliferation , Chlorocebus aethiops , Disulfides/chemistry , Humans , Mice , Molecular Sequence Data , Mutation , Protein Isoforms , Protein Structure, Tertiary , Receptors, Interleukin-3/chemistry , Signal Transduction
7.
J Biol Chem ; 285(29): 22370-81, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20472554

ABSTRACT

The cytokine interleukin-3 (IL-3) is a critical regulator of inflammation and immune responses in mammals. IL-3 exerts its effects on target cells via receptors comprising an IL-3-specific alpha-subunit and common beta-subunit (beta c; shared with IL-5 and granulocyte-macrophage colony-stimulating factor) or a beta-subunit that specifically binds IL-3 (beta(IL-3); present in mice but not humans). We recently identified two splice variants of the alpha-subunit of the IL-3 receptor (IL-3R alpha) that are relevant to hematopoietic progenitor cell differentiation or proliferation: the full length ("SP1" isoform) and a novel isoform (denoted "SP2") lacking the N-terminal Ig-like domain. Although our studies demonstrated that each mouse IL-3 (mIL-3) R alpha isoform can direct mIL-3 binding to two distinct sites on the beta(IL-3) subunit, it has remained unclear which residues in mIL-3 itself are critical to the two modes of beta(IL-3) recognition and whether the human IL-3R alpha SP1 and SP2 orthologs similarly instruct human IL-3 binding to two distinct sites on the human beta c subunit. Herein, we describe the identification of residues clustering around the highly conserved A-helix residue, Glu(23), in the mIL-3 A- and C-helices as critical for receptor binding and growth stimulation via the beta(IL-3) and mIL-3R alpha SP2 subunits, whereas an overlapping cluster was required for binding and activation of beta(IL-3) in the presence of mIL-3R alpha SP1. Similarly, our studies of human IL-3 indicate that two different modes of beta c binding are utilized in the presence of the hIL-3R alpha SP1 or SP2 isoforms, suggesting a possible conserved mechanism by which the relative orientations of receptor subunits are modulated to achieve distinct signaling outcomes.


Subject(s)
Cytokine Receptor Common beta Subunit/metabolism , Epitopes/metabolism , Interleukin-3/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cell Proliferation , Cytokine Receptor Common beta Subunit/chemistry , DNA Mutational Analysis , Enzyme Activation , Glutamic Acid/metabolism , Humans , Interleukin-3/chemistry , Interleukin-3 Receptor alpha Subunit/metabolism , Janus Kinase 2/metabolism , MAP Kinase Signaling System , Mice , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation/genetics , Protein Isoforms/chemistry , Protein Isoforms/metabolism
8.
Biochem J ; 426(3): 307-17, 2010 Feb 24.
Article in English | MEDLINE | ID: mdl-20078425

ABSTRACT

GM-CSF (granulocyte/macrophage colony-stimulating factor) is an important mediator of inducible haemopoiesis and inflammation, and has a critical role in the function of alveolar macrophages. Its clinical applications include the mobilization of haemopoietic progenitors, and a role as an immune stimulant and vaccine adjuvant in cancer patients. GM-CSF signals via a specific alpha receptor (GM-CSFRalpha) and the shared hbetac (human common beta-subunit). The present study has investigated the role of the Ig-like domain of GM-CSFRalpha in GM-CSF binding and signalling. Deletion of the Ig-like domain abolished direct GM-CSF binding and decreased growth signalling in the presence of hbetac. To locate the specific residues in the Ig-like domain of GM-CSFRalpha involved in GM-CSF binding, a structural alignment was made with a related receptor, IL-13Ralpha1 (interleukin-13 receptor alpha1), whose structure and mode of interaction with its ligand has recently been elucidated. Mutagenesis of candidate residues in the predicted region of interaction identified Val51 and Cys60 as having critical roles in binding to the alpha receptor, with Arg54 and Leu55 also being important. High-affinity binding in the presence of hbetac was strongly affected by mutation of Cys60 and was also reduced by mutation of Val51, Arg54 and Leu55. Of the four key residues, growth signalling was most severely affected by mutation of Cys60. The results indicate a previously unrecognized role for the Ig-like domain, and in particular Cys60, of GM-CSFRalpha in the binding of GM-CSF and subsequent activation of cellular signalling.


Subject(s)
Cytokines/metabolism , Receptors, Cytokine/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Binding Sites/genetics , COS Cells , Cell Line , Cell Proliferation , Chlorocebus aethiops , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Cytokine Receptor Common beta Subunit/chemistry , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/metabolism , Cytokines/genetics , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-13 Receptor alpha1 Subunit/chemistry , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-13 Receptor alpha1 Subunit/metabolism , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Structure, Tertiary , Receptors, Cytokine/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Sequence Homology, Amino Acid
9.
Growth Factors ; 28(2): 104-10, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19961363

ABSTRACT

Mouse interleukin-3 (mIL-3) is a critical cytokine regulator of myeloid cell differentiation, survival and activation, and consequently this cytokine has become a key reagent for hematological studies in the laboratory. Although bacterial expression has been used for the preparation of recombinant mIL-3 for more than 20 years, the resultant cytokine is known to exhibit poor solubility, be prone to aggregation, and may contain mispaired disulfide bonds. As a result, little structural characterization of mIL-3 has been possible to date. In the present work, we describe a convenient, inexpensive, and scalable protocol for preparing an mIL-3 analog with wild-type bioactivity from Escherichia coli via a simple purification scheme. This analog is typically expressed at >1 mg/l of shaking Super broth culture and, owing to solubility >5 mg/ml, structural studies in solution by nuclear magnetic resonance spectroscopy are feasible for mIL-3 for the first time.


Subject(s)
Escherichia coli/metabolism , Interleukin-3 , Protein Engineering/methods , Animals , Base Sequence , Cytokines/genetics , Cytokines/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Interleukin-3/chemistry , Interleukin-3/genetics , Interleukin-3/isolation & purification , Interleukin-3/metabolism , Magnetic Resonance Spectroscopy , Mice , Mutagenesis, Site-Directed , Solubility
10.
J Biol Chem ; 284(9): 5763-73, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19109256

ABSTRACT

Interleukin-3 (IL-3) promotes both self-renewal and differentiation of early multipotential progenitors and is involved in inducible hematopoiesis in response to infections. Here we report new insights into these processes with the identification of a new isoform (SP2) of IL-3 receptor alpha (IL-3Ralpha), present in mouse and human hematopoietic cells, which lacks domain 1 of the full-length receptor (SP1). Binding assays with beta(IL-3) mutants showed that mouse SP2 uses a different high affinity binding mode to SP1, although both mouse and human SP2 and SP1 can stimulate IL-3-dependent growth. In IL-3-dependent differentiation models, human SP2 and SP1 gave differential effects on lineage commitment or self-renewal dependent on the cellular context, suggesting that different modes of ectodomain binding may modulate intracellular signaling. In a multipotential factor dependent cell-Paterson mix, the transcription factors C/EBPalpha and PU.1 and microRNAs miRNA-15a, -223, and -181a were up-regulated in cells undergoing SP2-supported differentiation compared with SP1-supported self-renewal. Similarly in M1 cells, SP2 promoted differentiation compared with SP1 and gave up-regulation of PU.1 and miRNA-155 and -223. These findings suggest that IL-3-promoted lineage commitment uses similar mechanisms to those of steady-state hematopoiesis. Both the SP1 and SP2 isoforms activated the Jak2/STAT5, Akt, and Erk1/2 signaling pathways in M1 cells, although the activation was more prolonged for the SP2 isoform.


Subject(s)
Alternative Splicing , Cell Differentiation , Interleukin-3 Receptor alpha Subunit/metabolism , Receptors, Interleukin-3/metabolism , Signal Transduction , Animals , Blotting, Western , COS Cells , Cell Proliferation , Cells, Cultured , Chlorocebus aethiops , Flow Cytometry , Glycosylation , Humans , Interleukin-3 Receptor alpha Subunit/genetics , Janus Kinase 2/metabolism , Mice , Mice, Transgenic , MicroRNAs/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Isoforms , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin-3/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/metabolism , Transcription Factors/metabolism
11.
Cytokine ; 42(2): 234-242, 2008 May.
Article in English | MEDLINE | ID: mdl-18374598

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-3 and IL-5 are related cytokines that play key roles in regulating the differentiation, proliferation, survival and activation of myeloid blood cells. The cell surface receptors for these cytokines are composed of cytokine-specific alpha-subunits and a common beta-receptor (betac), a shared subunit that is essential for receptor signaling in response to GM-CSF, IL-3 and IL-5. Previous studies have reached conflicting conclusions as to whether N-glycosylation of the betac-subunit is necessary for functional GM-CSF, IL-3 and IL-5 receptors. We sought to clarify whether betac N-glycosylation plays a role in receptor function, since all structural studies of human betac to date have utilized recombinant protein lacking N-glycosylation at Asn(328). Here, by eliminating individual N-glycans in human betac and the related murine homolog, beta(IL-3), we demonstrate unequivocally that ligand-binding and receptor activation are not critically dependent on individual N-glycosylation sites within the beta-subunit although the data do not preclude the possibility that N-glycans may exert some sort of fine control. These studies support the biological relevance of the X-ray crystal structures of the human betac domain 4 and the complete ectodomain, both of which lack N-glycosylation at Asn(328).


Subject(s)
Cytokine Receptor Common beta Subunit/physiology , Polysaccharides/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Receptors, Interleukin-3/physiology , Receptors, Interleukin-5/physiology , Animals , COS Cells , Chlorocebus aethiops , Cytokine Receptor Common beta Subunit/chemistry , Cytokine Receptor Common beta Subunit/genetics , Humans , Interleukin-3/metabolism , Interleukin-5/metabolism , Mice , Mutagenesis, Site-Directed , Polysaccharides/chemistry , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Receptors, Interleukin-3/chemistry , Receptors, Interleukin-5/chemistry
12.
J Neurochem ; 100(3): 693-707, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17116230

ABSTRACT

Homologues of the Drosophila melanogaster tweety (tty) gene are present in mammals and Caenorhabditis elegans. The encoded proteins have five predicted membrane-spanning regions and recent findings suggest that some family members may be chloride channels. Phylogenetic analysis of the tty family including novel members from slime mould Entamoeba and plants has revealed the occurrence of independent gene duplication events in different lineages. expressed sequence tag data indicate that expression of the mammalian Ttyh1 gene is restricted mainly to neural tissue and is up-regulated in astrocytoma, glioma and several other cancers. In this study, mammalian expression vectors were used to investigate the subcellular localization and the effect of over-expression of Ttyh1 in human epithelial kidney cells. The results confirm that Ttyh1 is a membrane protein and show that it is deposited on the substratum along the migration paths of motile cells above the alpha5beta1-integrin complex. The ectopic expression of Ttyh1 also induced long filopodia, which were branched and dynamic in both stationary and migratory cells. The filopodia contained F-actin and occurred at the ends of microtubules which were polarized towards the membrane. Upon contact with nearby cells some filopodia stabilized and filled with F-actin, whereas Ttyh1 was highly concentrated at the cell-cell interface. Ttyh1 N- and C-terminal antipeptide antibodies detected Ttyh1 along the axons of neurones in primary rat hippocampal cell cultures, and in situ in whole rat brain slices around the hippocampus and occasionally between cells. These data suggest a role for Ttyh1 in process formation, cell adhesion and possibly as a transmembrane receptor.


Subject(s)
Brain/growth & development , Cell Membrane/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Actins/metabolism , Animals , Animals, Newborn , Brain/cytology , Brain/metabolism , Cell Adhesion/physiology , Cell Line , Cell Movement/physiology , Cell Polarity/physiology , Evolution, Molecular , Gene Expression Regulation, Developmental/genetics , Humans , Integrin alpha5/metabolism , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Neurons/cytology , Organ Culture Techniques , Pseudopodia/genetics , Pseudopodia/metabolism , Rats , Rats, Inbred BB , Rats, Long-Evans , Receptors, Cell Surface/metabolism
13.
Vitam Horm ; 74: 1-30, 2006.
Article in English | MEDLINE | ID: mdl-17027509

ABSTRACT

The cytokines, interleukin-3 (IL-3), interleukin-5 (IL-5), and granulocyte-macrophage colony stimulating factor (GM-CSF), are polypeptide growth factors that exhibit overlapping activities in the regulation of hematopoietic cells. They appear to be primarily involved in inducible hematopoiesis in response to infections and are involved in the pathogenesis of allergic and inflammatory diseases and possibly in leukemia. The X-ray structure of the beta common (betac) receptor ectodomain has given new insights into the structural biology of signaling by IL-3, IL-5, and GM-CSF. This receptor is shared between the three ligands and functions together with three ligand-specific alpha-subunits. The structure shows betac is an intertwined homodimer in which each chain contains four domains with approximate fibronectin type-III topology. The two betac-subunits that compose the homodimer are interlocked by virtue of the swapping of beta-strands between domain 1 of one subunit and domain 3 of the other subunit. Site-directed mutagenesis has shown that the interface between domains 1 and 4 in this unique structure forms the functional epitope. This epitope is similar to those of other members of the cytokine class I receptor family but is novel in that it is formed by two different receptor chains. The chapter also reviews knowledge on the closely related mouse beta(IL-3) receptor and on the alpha-subunit-ligand interactions. The knowledge on the two beta receptors is placed in context with advances in understanding of the structural biology of other members of the cytokine class I receptor family.


Subject(s)
Cytokine Receptor Common beta Subunit/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Interleukin-3/physiology , Interleukin-5/physiology , Signal Transduction/physiology , Animals , Crystallography, X-Ray , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Humans , Interleukin-3/chemistry , Interleukin-5/chemistry , Mice , Models, Molecular , Protein Binding , Protein Conformation
14.
Int Immunol ; 18(2): 313-23, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16373364

ABSTRACT

IL-5 is a key regulator of eosinophilic inflammation and is selectively expressed by antigen-activated Th2 lymphocytes. An important role for the proximal AP-1 and GATA sites in regulating IL-5 transcription is generally accepted but the significance of an adjacent Ets/NFAT site has remained unclear. We have investigated its role using the mouse Th2 clone D10.G4.1. Transcription of IL-5 reporter gene plasmids could be induced in D10 cells by phorbol myristate acetate/cyclic adenosine monophosphate (PMA/cAMP) stimulation and significantly further enhanced by activation of the mitogen-activated protein (MAP) kinase pathways. Strong induction of IL-5 mRNA was also induced by PMA/cAMP. Mutagenesis showed that the Ets/NFAT site is of critical importance along with the AP-1 and GATA sites in regulating IL-5 transcription stimulated by PMA/cAMP and MAP kinase activation. Transactivation was used to investigate the transcription factors which could function at the three sites and possible synergistic interactions. AP-1 (c-Fos/c-Jun) strongly induced IL-5 transcription and dominant negative AP-1 constructs confirmed that AP-1 plays an important role in regulating IL-5 expression. Ets1, unlike other members of the Ets/NFAT family, synergized strongly with AP-1 suggesting that Ets1 is the family member which functions at the Ets/NFAT site. AP-1/Ets1 transactivation also stimulated IL-5 mRNA expression. Ets1 binding to the proximal promoter region, demonstrated by chromatin immunoprecipitation, was stimulated by PMA/cAMP. The absolute dependence on the binding sites for Ets1, AP-1 and GATA-3 together with the strong synergy between Ets1 and AP-1 suggest close cooperative interactions between the three transcription factors in the regulation of IL-5 expression in mouse T cells.


Subject(s)
GATA3 Transcription Factor/physiology , Gene Expression Regulation , Interleukin-5/genetics , Proto-Oncogene Protein c-ets-1/physiology , Th2 Cells/metabolism , Transcription Factor AP-1/physiology , Animals , Binding Sites , Cyclic AMP/pharmacology , DNA , Genes, Reporter , Genes, fos , Genes, jun , MAP Kinase Signaling System/physiology , Mice , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , Protein Binding , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic , Transcriptional Activation
15.
J Biol Chem ; 279(25): 26500-8, 2004 Jun 18.
Article in English | MEDLINE | ID: mdl-15060062

ABSTRACT

Interleukin-3 (IL-3) is a cytokine produced by activated T-cells and mast cells that is active on a broad range of hematopoietic cells and in the nervous system and appears to be important in several chronic inflammatory diseases. In this study, alanine substitutions were used to investigate the role of residues of the human beta-common (hbetac) receptor and the murine IL-3-specific (beta(IL-3)) receptor in IL-3 binding. We show that the domain 1 residues, Tyr(15) and Phe(79), of the hbetac receptor are important for high affinity IL-3 binding and receptor activation as shown previously for the related cytokines, interleukin-5 and granulocyte-macrophage colony-stimulating factor, which also signal through this receptor subunit. From the x-ray structure of hbetac, it is clear that the domain 1 residues cooperate with domain 4 residues to form a novel ligand-binding interface involving the two protein chains of the intertwined homodimer receptor. We demonstrate by ultracentrifugation that the beta(IL-3) receptor is also a homodimer. Its high sequence homology with hbetac suggests that their structures are homologous, and we identified an analogous binding interface in beta(IL-3) for direct IL-3 binding to the high affinity binding site in hbetac. Tyr(21) (A-B loop), Phe(85), and Asn(87) (E-F loop) of domain 1; Ile(320) of the interdomain loop; and Tyr(348) (B'-C' loop) and Tyr(401) (F'-G' loop) of domain 4 were shown to have critical individual roles and Arg(84) and Tyr(317) major secondary roles in direct murine IL-3 binding to the beta(IL-3)receptor. Most surprising, none of the key residues for direct IL-3 binding were critical for high affinity binding in the presence of the murine IL-3 alpha receptor, indicating a fundamentally different mechanism of high affinity binding to that used by hbetac.


Subject(s)
Interleukin-3/metabolism , Receptors, Cell Surface/chemistry , Alanine/chemistry , Amino Acid Sequence , Animals , COS Cells , Cell Division , Cross-Linking Reagents/pharmacology , Crystallography, X-Ray , Cytokine Receptor Common beta Subunit , Cytokines/metabolism , DNA, Complementary/metabolism , Dimerization , Electrophoresis, Polyacrylamide Gel , Epitopes , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-5/metabolism , Kinetics , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Structure, Tertiary , Receptors, Cell Surface/metabolism , Sequence Homology, Amino Acid , Transfection , Ultracentrifugation
16.
J Allergy Clin Immunol ; 112(5): 935-43, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14610483

ABSTRACT

BACKGROUND: Eotaxin-2 is a member of the eotaxin subfamily of CC chemokines that display eosinophil-specific, chemotactic properties and has been associated with allergic disorders. However, the contribution of eotaxin-2 to the development of defined pathogenic features of allergic disease remains to be defined. OBJECTIVE: We sought to determine whether eotaxin-2 was a cofactor with IL-5 for the regulation of pulmonary eosinophilia and to identify the combined role of these molecules in the induction of phenotypic characteristics of allergic lung disease. METHODS: We instilled recombinant eotaxin-2 into the airways of wild-type mice that had been treated systemically with IL-5 or into IL-5-transgenic mice and characterized pulmonary eosinophil numbers, IL-13 production, and airway hyperreactivity (AHR) to methacholine. Mice deficient in the IL-4 receptor alpha-chain, IL-13, and signal transducers and activators of transcription 6 or mice treated with anti-CCR3 monoclonal antibody were also used. RESULTS: Eotaxin-2 and IL-5 cooperatively promoted eosinophil accumulation, IL-13 production, and AHR to methacholine. Neither eotaxin-2 nor IL-5 alone induced these features of allergic disease. IL-13 production was critically dependent on eotaxin-2- and IL-5-regulated eosinophilia, which predisposed to the development of AHR. AHR was dependent on IL-13 and signaling through the IL-4R alpha-chain and signal transducers and activators of transcription 6 pathways and the presence of eosinophils in the lung. CONCLUSION: These investigations demonstrate important cooperativity between eotaxin-2, IL-5, and IL-13 signaling systems and eosinophils for the development of hallmark features of allergic disease of the lung.


Subject(s)
Bronchial Hyperreactivity/chemically induced , Chemokines, CC , Interleukin-13/biosynthesis , Interleukin-5 , Lung/metabolism , Pulmonary Eosinophilia/chemically induced , Aerosols , Animals , Bronchial Hyperreactivity/physiopathology , Bronchoconstrictor Agents/administration & dosage , Chemokine CCL24 , Chemokines, CC/administration & dosage , Drug Synergism , Eosinophils/pathology , Instillation, Drug , Leukocyte Count , Lung/drug effects , Methacholine Chloride/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Knockout/genetics , Mice, Transgenic/genetics , Pulmonary Eosinophilia/pathology , Receptors, Interleukin-4/genetics , Receptors, Interleukin-4/metabolism , Recombinant Proteins/administration & dosage , STAT6 Transcription Factor , Trans-Activators/deficiency , Trans-Activators/metabolism
17.
J Biol Chem ; 278(12): 10572-7, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12525483

ABSTRACT

The receptors for human interleukins 3 and 5 and granulocyte macrophage colony-stimulating factor are composed of ligand-specific alpha-subunits and a common beta-subunit (betac), the major signaling entity. The way in which betac interacts with ligands in the respective activation complexes has remained poorly understood. The recently determined crystal structure of the extracellular domain of betac revealed a possible ligand-binding interface composed of domain 1 of one chain of the betac dimer and the adjacent domain 4 of the symmetry-related chain. We have used site-directed mutagenesis, in conjunction with ligand binding and proliferation studies, to demonstrate the critical requirement of the domain 1 residues, Tyr(15) (A-B loop) and Phe(79) (E-F loop), in high affinity complex formation and receptor activation. The novel ligand-receptor interface formed between domains 1 and 4 represents the first example of a class I cytokine receptor interface to be composed of two noncontiguous fibronectin III domains.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interleukin-5/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Receptors, Interleukin-3/chemistry , Receptors, Interleukin/chemistry , Epitopes , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-5/metabolism , Protein Subunits , Receptors, Interleukin-5 , Structure-Activity Relationship
18.
J Exp Med ; 195(11): 1433-44, 2002 Jun 03.
Article in English | MEDLINE | ID: mdl-12045241

ABSTRACT

Interleukin (IL)-5 and IL-13 are thought to play key roles in the pathogenesis of asthma. Although both cytokines use eotaxin to regulate eosinophilia, IL-13 is thought to operate a separate pathway to IL-5 to induce airways hyperreactivity (AHR) in the allergic lung. However, identification of the key pathway(s) used by IL-5 and IL-13 in the disease process is confounded by the failure of anti-IL-5 or anti-IL-13 treatments to completely inhibit the accumulation of eosinophils in lung tissue. By using mice deficient in both IL-5 and eotaxin (IL-5/eotaxin(-/-)) we have abolished tissue eosinophilia and the induction of AHR in the allergic lung. Notably, in mice deficient in IL-5/eotaxin the ability of CD4(+) T helper cell (Th)2 lymphocytes to produce IL-13, a critical regulator of airways smooth muscle constriction and obstruction, was significantly impaired. Moreover, the transfer of eosinophils to IL-5/eotaxin(-/-) mice overcame the intrinsic defect in T cell IL-13 production. Thus, factors produced by eosinophils may either directly or indirectly modulate the production of IL-13 during Th2 cell development. Our data show that IL-5 and eotaxin intrinsically modulate IL-13 production from Th2 cells and that these signaling systems are not necessarily independent effector pathways and may also be integrated to regulate aspects of allergic disease.


Subject(s)
Asthma/complications , Bronchial Hyperreactivity/metabolism , CD4-Positive T-Lymphocytes/metabolism , Chemokines, CC/metabolism , Eosinophilia/metabolism , Interleukin-13/metabolism , Interleukin-5/metabolism , Adoptive Transfer , Animals , Asthma/immunology , Asthma/metabolism , Bronchial Hyperreactivity/complications , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , CD4-Positive T-Lymphocytes/immunology , Chemokine CCL11 , Chemokines, CC/blood , Chemokines, CC/genetics , Disease Models, Animal , Eosinophilia/complications , Eosinophilia/immunology , Eosinophilia/pathology , Eosinophils/metabolism , Eosinophils/transplantation , Gene Deletion , Humans , Interleukin-13/biosynthesis , Interleukin-18/metabolism , Interleukin-5/blood , Interleukin-5/genetics , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Sputum/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
19.
Mol Cell Biol ; 22(10): 3518-26, 2002 May.
Article in English | MEDLINE | ID: mdl-11971982

ABSTRACT

The Drosophila melanogaster flightless I gene is required for normal cellularization of the syncytial blastoderm. Highly conserved homologues of flightless I are present in Caenorhabditis elegans, mouse, and human. We have disrupted the mouse homologue Fliih by homologous recombination in embryonic stem cells. Heterozygous Fliih mutant mice develop normally, although the level of Fliih protein is reduced. Cultured homozygous Fliih mutant blastocysts hatch, attach, and form an outgrowing trophoblast cell layer, but egg cylinder formation fails and the embryos degenerate. Similarly, Fliih mutant embryos initiate implantation in vivo but then rapidly degenerate. We have constructed a transgenic mouse carrying the complete human FLII gene and shown that the FLII transgene is capable of rescuing the embryonic lethality of the homozygous targeted Fliih mutation. These results confirm the specific inactivation of the Fliih gene and establish that the human FLII gene and its gene product are functional in the mouse. The Fliih mouse mutant phenotype is much more severe than in the case of the related gelsolin family members gelsolin, villin, and CapG, where the homozygous mutant mice are viable and fertile but display alterations in cytoskeletal actin regulation.


Subject(s)
Cytoskeleton/metabolism , Drosophila Proteins , Embryo, Mammalian/physiology , Gelsolin , Proteins/metabolism , Stem Cells/physiology , Actins/metabolism , Animals , Carrier Proteins , Cells, Cultured , Cytoskeletal Proteins , Embryo Implantation , Embryo, Mammalian/anatomy & histology , Female , Gene Targeting , Humans , Insect Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Microfilament Proteins , Pregnancy , Stem Cells/cytology , Trans-Activators , Uterus/cytology
20.
Trends Mol Med ; 8(4): 162-7, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11927273

ABSTRACT

Extensive clinical investigations have implicated eosinophils in the pathogenesis of asthma. In a recent clinical trial, humanized monoclonal antibody to interleukin (IL)-5 significantly limited eosinophil migration to the lung. However, treatment did not affect the development of the late-phase response or airways hyperresponsiveness in experimental asthma. Although IL-5 is a key regulator of eosinophilia and attenuation of its actions without signs of clinical improvement raises questions about the contribution of these cells to disease, further studies are warranted to define the effects of anti-IL-5 in the processes that lead to chronic asthma. Furthermore, eosinophil accumulation into allergic tissues should not be viewed as a process that is exclusively regulated by IL-5 but one in which IL-5 greatly contributes. Indeed, data on anti-IL-5 treatments (human and animal models) are confounded by the failure of this approach to completely resolve tissue eosinophilia and the belief that IL-5 alone is the critical molecular switch for eosinophil development and migration. The contribution of these IL-5-independent pathways should be considered when assessing the role of eosinophils in disease processes.


Subject(s)
Asthma/immunology , Asthma/therapy , Eosinophils/metabolism , Interleukin-5/metabolism , Animals , Asthma/physiopathology , Bronchial Hyperreactivity , Disease Models, Animal , Eosinophilia , Eosinophils/immunology , Humans , Interleukin-5/immunology , Lung/immunology , Lung/pathology , Lung/physiology , Lung/physiopathology , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...