Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 307
Filter
1.
Curr Top Microbiol Immunol ; 316: 23-37, 2007.
Article in English | MEDLINE | ID: mdl-17969442

ABSTRACT

Interferon alpha (IFN-alpha) was approved by the US Food and Drug Administration on June 5, 1986 and paved the way for development of many other cytokines and growth factors. Nevertheless, we have barely touched the surface of understanding the multitude of human IFNs. This paper reviews the history of the purification of human leukocyte IFN, the cloning of the IFN-alphas, and the current state of knowledge of human interferon alpha genes and proteins.


Subject(s)
Interferon-alpha/genetics , Interferon-alpha/isolation & purification , Chromatography, High Pressure Liquid , Cloning, Molecular , DNA/chemistry , DNA/genetics , History, 20th Century , Humans , Interferon-alpha/chemistry , Interferon-alpha/history
2.
J Immunol ; 167(12): 6884-92, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11739506

ABSTRACT

In this study, we characterized the differential receptor-binding specificity, affinity, and Janus kinase-STAT activation of cellular IL-10 (cIL-10) compared with viral IL-10 (vIL-10). Only cells expressing IL-10R1 bind human IL-10 or vIL-10. IL-10R2 does not bind to cIL-10 or vIL-10 alone and its presence does not enhance the receptor-binding affinity of cIL-10 or vIL-10, but it is essential for both cIL-10- and vIL-10-mediated signal transduction and immune regulation. Responses initiated by cIL-10 and vIL-10 were compared in B cell and mast cell lines, and demonstrated that the inability of vIL-10 to stimulate immune responses, as compared with human IL-10, is due to failure to initiate signaling. Absent signal transduction is due to low level expression of cell surface IL-10R1, since overexpressing IL-10R1 allows vIL-10 to initiate cIL-10-like signals and subsequent biological responses. These results are similar in primary cells, since splenocytes respond to both cIL-10 and vIL-10, while thymocytes respond only to cIL-10 and have very low mouse IL-10R1 but not mouse IL-10R2 expression. These data demonstrate that IL-10R1 expression plays a critical role in determining whether cells respond to IL-10. Modulation of cell surface IL-10R1 density might be an important mechanism for determining whether IL-10 leads to immunostimulation or immunosuppression in vivo.


Subject(s)
Interleukin-10/pharmacology , Milk Proteins , Receptors, Interleukin/metabolism , Receptors, Interleukin/physiology , Signal Transduction , Animals , B-Lymphocytes/immunology , CHO Cells , Cell Line , Cells, Cultured , Cricetinae , DNA-Binding Proteins/metabolism , Humans , Interleukin-10/metabolism , Lymphocyte Activation , Mast Cells/immunology , Mice , Protein Isoforms/physiology , RNA, Messenger/biosynthesis , Receptors, Interleukin/genetics , Receptors, Interleukin-10 , STAT1 Transcription Factor , STAT3 Transcription Factor , STAT5 Transcription Factor , T-Lymphocytes/immunology , Trans-Activators/metabolism , Transcription, Genetic , Viral Proteins/pharmacology
3.
J Leukoc Biol ; 70(6): 950-60, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11739558

ABSTRACT

The heterodimeric interferon (IFN)-gamma receptor (IFN-gammaR) is formed of two chains. Here we show that the binding chain (IFN-gammaR1) was highly expressed on the membranes of T, B, and myeloid cells. Conversely, the transducing chain (IFN-gammaR2) was highly expressed on the surfaces of myeloid cells, moderately expressed on B cells, and poorly expressed on the surfaces of T cells. Differential cell membrane expression of IFN-gammaR2 determined the number of receptor complexes that transduced the IFN-gamma signal and resulted in a different response to IFN-gamma. After IFN-gamma stimulation, high IFN-gammaR2 membrane expression induced rapid activation of signal transducer and activator of transcription-1 (STAT-1) and high levels of interferon regulatory factor-1 (IRF-1), which then triggered the apoptotic program. By contrast, low cell membrane expression resulted in slow activation of STAT-1, lower levels of IRF-1, and induction of proliferation. Because the forced expression of IFN-gammaR2 on T cells switched their response to IFN-gamma from proliferative to apoptotic, we concluded that the surface expression of IFN-gammaR2 determines whether a cell stimulated by IFN-gamma undergoes proliferation or apoptosis.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/immunology , Myeloid Cells/immunology , Receptors, Interferon/immunology , T-Lymphocytes/immunology , B-Lymphocytes/cytology , Cell Division/immunology , Cells, Cultured , DNA-Binding Proteins/immunology , Humans , Interferon Regulatory Factor-1 , Interferon-gamma/immunology , Myeloid Cells/cytology , Phosphoproteins/immunology , STAT1 Transcription Factor , Signal Transduction/immunology , T-Lymphocytes/cytology , Trans-Activators/immunology , Interferon gamma Receptor
4.
Oncogene ; 20(48): 7051-63, 2001 Oct 25.
Article in English | MEDLINE | ID: mdl-11704829

ABSTRACT

Abnormalities in cellular differentiation are frequent occurrences in human cancers. Treatment of human melanoma cells with recombinant fibroblast interferon (IFN-beta) and the protein kinase C activator mezerein (MEZ) results in an irreversible loss in growth potential, suppression of tumorigenic properties and induction of terminal cell differentiation. Subtraction hybridization identified melanoma differentiation associated gene-7 (mda-7), as a gene induced during these physiological changes in human melanoma cells. Ectopic expression of mda-7 by means of a replication defective adenovirus results in growth suppression and induction of apoptosis in a broad spectrum of additional cancers, including melanoma, glioblastoma multiforme, osteosarcoma and carcinomas of the breast, cervix, colon, lung, nasopharynx and prostate. In contrast, no apparent harmful effects occur when mda-7 is expressed in normal epithelial or fibroblast cells. Human clones of mda-7 were isolated and its organization resolved in terms of intron/exon structure and chromosomal localization. Hu-mda-7 encompasses seven exons and six introns and encodes a protein with a predicted size of 23.8 kDa, consisting of 206 amino acids. Hu-mda-7 mRNA is stably expressed in the thymus, spleen and peripheral blood leukocytes. De novo mda-7 mRNA expression is also detected in human melanocytes and expression is inducible in cells of melanocyte/melanoma lineage and in certain normal and cancer cell types following treatment with a combination of IFN-beta plus MEZ. Mda-7 expression is also induced during megakaryocyte differentiation induced in human hematopoietic cells by treatment with TPA (12-O-tetradecanoyl phorbol-13-acetate). In contrast, de novo expression of mda-7 is not detected nor is it inducible by IFN-beta+MEZ in a spectrum of additional normal and cancer cells. No correlation was observed between induction of mda-7 mRNA expression and growth suppression following treatment with IFN-beta+MEZ and induction of endogenous mda-7 mRNA by combination treatment did not result in significant intracellular MDA-7 protein. Radiation hybrid mapping assigned the mda-7 gene to human chromosome 1q, at 1q 32.2 to 1q41, an area containing a cluster of genes associated with the IL-10 family of cytokines. Mda-7 represents a differentiation, growth and apoptosis associated gene with potential utility for the gene-based therapy of diverse human cancers.


Subject(s)
Antigens, Neoplasm/genetics , Apoptosis/genetics , Chromosomes, Human, Pair 1/genetics , Diterpenes , Genes , Growth Substances/genetics , Interleukins , Neoplasm Proteins/genetics , Neoplasms/genetics , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/isolation & purification , Base Sequence , Carcinoma/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Division/genetics , Cloning, Molecular , Dimethyl Sulfoxide/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor , Glioblastoma/pathology , Growth Substances/biosynthesis , Growth Substances/isolation & purification , HL-60 Cells/metabolism , HL-60 Cells/pathology , Humans , Interferon Type I/pharmacology , K562 Cells/metabolism , K562 Cells/pathology , Male , Melanocytes/metabolism , Melanoma/chemistry , Melanoma/genetics , Melanoma/pathology , Molecular Sequence Data , Molecular Weight , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/isolation & purification , Organ Specificity , Osteosarcoma/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Recombinant Fusion Proteins/physiology , Recombinant Proteins , Terpenes/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Tumor Cells, Cultured/pathology
5.
J Biol Chem ; 276(52): 49034-42, 2001 Dec 28.
Article in English | MEDLINE | ID: mdl-11679576

ABSTRACT

The Jak family of protein-tyrosine kinases are crucial for the signaling of a large number of different polypeptide ligands, including the interferons, many cytokines, erythropoietin, and growth factors. Through their interaction with receptors, the Jaks initiate a signaling cascade resulting in the activation of gene transcription and ultimately a cellular response to various ligands. In addition to their role in cellular signaling, alteration of Jak activity has been implicated in several disease states. In identifying Jak2-interacting proteins with the yeast two-hybrid system, we cloned the human homologue of the Drosophila melanogaster tumor suppressor gene lethal () tumorous imaginal discs, which encodes the protein Tid56. Drosophila Tid56 and its human homologue hTid-1 represent members of the DnaJ family of molecular chaperones. The TID1 gene encodes two splice variants hTid-1(S) and hTid-1(L). We confirmed the interaction between Jak2 and hTid-1(S) or hTid-1(L) by immunoprecipitation from COS-1 cells expressing these proteins. The interaction between endogenous hTid-1 and Jak2 was shown in HEp2 cells. We further showed that hTid-1 interacts with the human interferon-gamma (Hu-IFN-gamma) receptor subunit IFN-gamma R2. In addition, using a chimeric construct where the extracellular domain of IFN-gamma R2 was fused to the kinase domain of Jak2, we showed that hTid-1 binds more efficiently to the chimera with an active kinase domain than to a similar construct with an inactive kinase domain. Additionally, the data demonstrate that hTid-1 isoforms as well as Jak2 interact with Hsp70/Hsc70 in vivo, and the interaction between Hsp70/Hsc70 and hTid-1 is reduced after IFN-gamma treatment. Furthermore, both hTid-1(S) and hTid-1(L) can modulate IFN-gamma-mediated transcriptional activity.


Subject(s)
Heat-Shock Proteins/metabolism , Interferon-gamma/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins , Signal Transduction/physiology , Animals , COS Cells , Cell Fractionation , Genes, Reporter , HSP40 Heat-Shock Proteins , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Humans , Janus Kinase 2 , Models, Biological , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Two-Hybrid System Techniques
6.
J Immunol ; 166(12): 7096-103, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11390454

ABSTRACT

With the use of a partial sequence of the human genome, we identified a gene encoding a novel soluble receptor belonging to the class II cytokine receptor family. This gene is positioned on chromosome 6 in the vicinity of the IFNGR1 gene in a head-to-tail orientation. The gene consists of six exons and encodes a 231-aa protein with a 21-aa leader sequence. The secreted mature protein demonstrates 34% amino acid identity to the extracellular domain of the IL-22R1 chain. Cross-linking experiments demonstrate that the protein binds IL-22 and prevents binding of IL-22 to the functional cell surface IL-22R complex, which consists of two subunits, the IL-22R1 and the IL-10R2c chains. Moreover, this soluble receptor, designated IL-22-binding protein (BP), is capable of neutralizing IL-22 activity. In the presence of the IL-22BP, IL-22 is unable to induce Stat activation in IL-22-responsive human lung carcinoma A549 cells. IL-22BP also blocked induction of the suppressors of cytokine signaling-3 (SOCS-3) gene expression by IL-22 in HepG2 cells. To further evaluate IL-22BP action, we used hamster cells expressing a modified IL-22R complex consisting of the intact IL-10R2c and the chimeric IL-22R1/gammaR1 receptor in which the IL-22R1 intracellular domain was replaced with the IFN-gammaR1 intracellular domain. In these cells, IL-22 activates biological activities specific for IFN-gamma, such as up-regulation of MHC class I Ag expression. The addition of IL-22BP neutralizes the ability of IL-22 to induce Stat activation and MHC class I Ag expression in these cells. Thus, the soluble receptor designated IL-22BP inhibits IL-22 activity by binding IL-22 and blocking its interaction with the cell surface IL-22R complex.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/genetics , Interleukins/antagonists & inhibitors , Interleukins/metabolism , Receptors, Cell Surface , Amino Acid Sequence , Animals , Base Sequence , Binding, Competitive/immunology , CHO Cells , COS Cells , Carrier Proteins/biosynthesis , Carrier Proteins/metabolism , Carrier Proteins/physiology , Cloning, Molecular , Cricetinae , DNA, Complementary/isolation & purification , Humans , Ligands , Molecular Sequence Data , Receptors, Interleukin , Solubility , Tumor Cells, Cultured , Interleukin-22
7.
J Biol Chem ; 276(35): 32971-6, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11413150

ABSTRACT

We have identified a new mammalian protein arginine N-methyltransferase, PRMT5, formerly designated Janus kinase-binding protein 1, that can catalyze the formation of omega-N(G)-monomethylarginine and symmetric omega-N(G),N(G')-dimethylarginine in a variety of proteins. A hemagglutinin peptide-tagged PRMT5 complex purified from human HeLa cells catalyzes the S-adenosyl-l-[methyl-(3)H]methionine-dependent in vitro methylation of myelin basic protein. When the radiolabeled myelin basic protein was acid-hydrolyzed to free amino acids, and the products were separated by high-resolution cation exchange chromatography, we were able to detect two tritiated species. One species co-migrated with a omega-N(G)-monomethylarginine standard, and the other co-chromatographed with a symmetric omega-N(G),N(G')-dimethylarginine standard. Upon base treatment, this second species formed methylamine, a breakdown product characteristic of symmetric omega-N(G),N(G')-dimethylarginine. Further analysis of these two species by thin layer chromatography confirmed their identification as omega-N(G)-monomethylarginine and symmetric omega-N(G),N(G')-dimethylarginine. The hemagglutinin-PRMT5 complex was also able to monomethylate and symmetrically dimethylate bovine histone H2A and a glutathione S-transferase-fibrillarin (amino acids 1-148) fusion protein (glutathione S-transferase-GAR). A mutation introduced into the S-adenosyl-l-methionine-binding motif I of a myc-tagged PRMT5 construct in COS-1 cells led to a near complete loss of observed enzymatic activity. PRMT5 is the first example of a catalytic chain for a type II protein arginine N-methyltransferase that can result in the formation of symmetric dimethylarginine residues as observed previously in myelin basic protein, Sm small nuclear ribonucleoproteins, and other polypeptides.


Subject(s)
Arginine/biosynthesis , Protein Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Arginine/analogs & derivatives , Arginine/chemistry , Binding Sites , HeLa Cells , Humans , Methylation , Models, Molecular , Myelin Basic Protein/metabolism , Protein Conformation , Protein Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , S-Adenosylmethionine/metabolism
8.
Gene ; 267(2): 233-42, 2001 Apr 18.
Article in English | MEDLINE | ID: mdl-11313150

ABSTRACT

Defects in growth control and differentiation occur frequently in human cancers. In the case of human melanoma cells, treatment with a combination of fibroblast interferon (IFN-beta) and the protein kinase C activator mezerein (MEZ) results in an irreversible loss of proliferative potential and tumorigenic properties with a concomitant induction of terminal differentiation. These changes in cellular properties are associated with an induction and suppression in specific subsets of genes that occur in a temporal manner. To identify the complete repertoire of gene changes occurring during melanoma reversion to a more differentiated state a number of molecular approaches are being used. These include, subtraction hybridization using temporally spaced cDNA libraries, random cDNA isolation and evaluation by reverse Northern blotting and high throughput microarray analysis of subtracted cDNA clones. In the present study we have used a novel approach, rapid subtraction hybridization (RaSH), to identify and clone an additional gene of potential relevance to cancer growth control and terminal cell differentiation. RaSH has identified a human ubiquitin-processing protease gene, HuUBP43, that is differentially expressed in melanoma cells as a function of treatment with IFN-beta or IFN-beta + MEZ. HuUBP43 is a type I interferon inducible gene that is upregulated in a diverse panel of normal and tumor cells when treated with IFN-beta via the JAK/STAT kinase pathway. This gene may contribute to the phenotypic changes induced by IFN-beta during growth arrest and differentiation in human melanoma cells and other cell types as well as the antiviral and growth inhibitory effects of interferon.


Subject(s)
Cloning, Molecular/methods , Diterpenes , Endopeptidases/genetics , Melanoma/genetics , Amino Acid Sequence , Base Sequence , Blotting, Northern , Cell Differentiation , Female , Gene Expression Regulation, Enzymologic/drug effects , HeLa Cells , Humans , Interferon-beta/pharmacology , Male , Melanoma/pathology , Molecular Sequence Data , Nucleic Acid Hybridization/methods , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/drug effects , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Terpenes/pharmacology , Tissue Distribution , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Ubiquitin Thiolesterase
9.
J Biol Chem ; 276(4): 2725-32, 2001 Jan 26.
Article in English | MEDLINE | ID: mdl-11035029

ABSTRACT

Interleukin-10 (IL-10)-related T cell-derived inducible factor (IL-TIF; provisionally designated IL-22) is a cytokine with limited homology to IL-10. We report here the identification of a functional IL-TIF receptor complex that consists of two receptor chains, the orphan CRF2-9 and IL-10R2, the second chain of the IL-10 receptor complex. Expression of the CRF2-9 chain in monkey COS cells renders them sensitive to IL-TIF. However, in hamster cells both chains, CRF2-9 and IL-10R2, must be expressed to assemble the functional IL-TIF receptor complex. The CRF2-9 chain (or the IL-TIF-R1 chain) is responsible for Stat recruitment. Substitution of the CRF2-9 intracellular domain with the IFN-gammaR1 intracellular domain changes the pattern of IL-TIF-induced Stat activation. The CRF2-9 gene is expressed in normal liver and kidney, suggesting a possible role for IL-TIF in regulating gene expression in these tissues. Each chain, CRF2-9 and IL-10R2, is capable of binding IL-TIF independently and can be cross-linked to the radiolabeled IL-TIF. However, binding of IL-TIF to the receptor complex is greater than binding to either receptor chain alone. Sharing of the common IL-10R2 chain between the IL-10 and IL-TIF receptor complexes is the first such case for receptor complexes with chains belonging to the class II cytokine receptor family, establishing a novel paradigm for IL-10-related ligands similar to the shared use of the gamma common chain (gamma(c)) by several cytokines, including IL-2, IL-4, IL-7, IL-9, and IL-15.


Subject(s)
Cytokines/metabolism , Interleukins/metabolism , Receptors, Interleukin/isolation & purification , Amino Acid Sequence , Cross-Linking Reagents , Humans , Ligands , Models, Biological , Molecular Sequence Data , Protein Binding , Receptors, Interleukin-10 , Signal Transduction , Interleukin-22
11.
Biochem Biophys Res Commun ; 274(1): 105-11, 2000 Jul 21.
Article in English | MEDLINE | ID: mdl-10903903

ABSTRACT

The yeast protein Hsl7p is a homologue of Janus kinase binding protein 1, JBP1, a newly characterized protein methyltransferase. In this report, Hsl7p also is shown to be a methyltransferase. It can be crosslinked to [(3)H]S-adenosylmethionine and exhibits in vitro protein methylation activity. Calf histones H2A and H4 and bovine myelin basic protein were methylated by Hsl7p, whereas histones H1, H2B, and H3 and bovine cytochrome c were not. We demonstrated that JBP1 can complement Saccharomyces cerevisiae with a disrupted HSL7 gene as judged by a reduction of the elongated bud phenotype, and a point mutation in the JBP1 S-adenosylmethionine consensus binding sequence eliminated all complementation by JBP1. Therefore, we conclude the yeast protein Hsl7p is a sequence and functional homologue of JBP1. These data provide evidence for an intricate link between protein methylation and macroscopic changes in yeast morphology.


Subject(s)
Protein Kinases/metabolism , Protein Methyltransferases/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/enzymology , Animals , Cattle , Cytochrome c Group/metabolism , Galactose/metabolism , Genetic Complementation Test , Histones/metabolism , Humans , Methylation , Methyltransferases/metabolism , Mutagenesis , Myelin Basic Protein/metabolism , Phenotype , Plasmids/metabolism , Point Mutation , Precipitin Tests , Protein Kinases/chemistry , Protein Kinases/genetics , Protein Methyltransferases/chemistry , Protein Methyltransferases/genetics , Protein Structure, Tertiary , Protein-Arginine N-Methyltransferases , S-Adenosylmethionine/metabolism , Ultraviolet Rays
12.
Oncogene ; 19(21): 2557-65, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10851054

ABSTRACT

Cells of the immune system communicate with each other to initiate, establish and maintain immune responses. The communication occurs through cell-to-cell contact or through a variety of intercellular mediators that include cytokines, chemokines, growth factors and hormones. In the case of cytokines, the signal is transmitted from the outside to the inside of a cell through cell surface receptors specific for each cytokine. At this step the signal is also decoded and amplified: ligand binding causes recruitment and/or activation of numerous cytoplasmic proteins. One cytokine can activate a number of signal transduction pathways leading to regulation of a wide array of biological activities. One of these pathways, the Jak-Stat pathway, is briefly reviewed here with respect to the class II cytokine receptors. Signal transduction through receptors for interferons Type I (IFN-alpha, IFN-beta, IFN-omega) and Type II (IFN-gamma), and interleukin 10 (IL-10) is described in detail. In addition, a complex between tissue factor (TF) and coagulation factor VIIa, and two new receptors related to the class II cytokine receptor family are discussed. Oncogene (2000).


Subject(s)
DNA-Binding Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Cytokine/metabolism , Signal Transduction , Trans-Activators/metabolism , Amino Acid Sequence , Animals , Humans , Janus Kinase 1 , Molecular Sequence Data , Receptors, Cytokine/chemistry , Receptors, Interferon/chemistry , Receptors, Interferon/metabolism , Receptors, Interleukin/metabolism , Receptors, Interleukin-10 , STAT1 Transcription Factor , Sequence Alignment
13.
J Biol Chem ; 275(30): 22995-3004, 2000 Jul 28.
Article in English | MEDLINE | ID: mdl-10811814

ABSTRACT

Interferon-gamma (IFN-gamma) and its receptor complex are dimeric and bilaterally symmetric. We created mutants of IFN-gamma that bind only one IFN-gammaR1 chain per dimer molecule (called a monovalent IFN-gamma) to see if the interaction of IFN-gamma with one-half of the receptor complex is sufficient for bioactivity. Mutating a receptor-binding sequence in either AB loop of a covalent dimer of IFN-gamma yielded two monovalent IFN-gammas, gamma(m)-gamma and gamma-gamma(m), which cross-link to only a single soluble IFN-gammaR1 molecule in solution and on the cell surface. Monovalent IFN-gamma competes fully with wild type IFN-gamma for binding to U937 cells but only at a greater than 100-fold higher concentration than wild type IFN-gamma. Monovalent IFN-gamma had anti-vesicular stomatitis virus activity and antiproliferative activity, and it induced major histocompatibility complex class I and class II (HLA-DR) expression. In contrast, the maximal levels of activated Stat1alpha produced by monovalent IFN-gammas after 15 min were never more than half of those produced by either wild type or covalent IFN-gammas in human cell lines. These data indicate that while monovalent IFN-gamma activates only one-half of a four-chain receptor complex, this is sufficient for Stat1alpha activation, major histocompatibility complex class I surface antigen induction, and antiviral and antiproliferative activities. Thus, while interaction with both halves of the receptor complex is required for high affinity binding of IFN-gamma and efficient signal transduction, interaction with only one-half of the receptor complex is sufficient to initiate signal transduction.


Subject(s)
Interferon-gamma/metabolism , Receptors, Interferon/metabolism , Signal Transduction , Base Sequence , Biopolymers , Cell Line , Chromatography, Gel , DNA Primers , Dimerization , Humans , Interferon-gamma/chemistry , Protein Binding , Interferon gamma Receptor
14.
Proc Natl Acad Sci U S A ; 97(4): 1695-700, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10677520

ABSTRACT

We identified a viral IL-10 homolog encoded by an ORF (UL111a) within the human cytomegalovirus (CMV) genome, which we designated cmvIL-10. cmvIL-10 can bind to the human IL-10 receptor and can compete with human IL-10 for binding sites, despite the fact that these two proteins are only 27% identical. cmvIL-10 requires both subunits of the IL-10 receptor complex to induce signal transduction events and biological activities. The structure of the cmvIL-10 gene is unique by itself. The gene retained two of four introns of the IL-10 gene, but the length of the introns was reduced. We demonstrated that cmvIL-10 is expressed in CMV-infected cells. Thus, expression of cmvIL-10 extends the range of counter measures developed by CMV to circumvent detection and destruction by the host immune system.


Subject(s)
Cytomegalovirus/genetics , Growth Substances/genetics , Interleukin-10/genetics , Viral Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , Cytomegalovirus/chemistry , DNA-Binding Proteins/metabolism , Genes, Viral/genetics , Genome, Viral , Growth Substances/chemistry , Humans , Interleukin-10/chemistry , Leukocytes , Major Histocompatibility Complex/genetics , Molecular Sequence Data , Protein Binding , Receptors, Interleukin/metabolism , Receptors, Interleukin-10 , STAT3 Transcription Factor , Sequence Alignment , Signal Transduction , Trans-Activators/metabolism , Transfection , Viral Proteins/chemistry
15.
J Immunol ; 164(7): 3506-11, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10725704

ABSTRACT

Activated T lymphocytes modulate the level of many molecules on their cell surface, including cytokine receptors. This regulation of cytokine receptor expression affects the ability of T cells to respond to cytokines and thus influences the outcome of an immune response. The receptor for IFN-gamma, a proinflammatory cytokine, consists of two copies of a ligand binding chain (IFN-gammaR1) as well as two copies of a second chain (IFN-gammaR2) required for signal transduction. The expression of IFN-gammaR2 is down-regulated at the mRNA level on CD4+ T cells when they differentiate into the Th1, but not the Th2, phenotype. This down-regulation has been demonstrated to depend on the ligand, IFN-gamma, which is produced by Th1 but not Th2 T cells. The regulation of the cell-surface expression of IFN-gamma receptors during primary T cell activation has not been reported. Naive and differentiated T lymphocytes express IFN-gammaR1 at the mRNA level and as a cell-surface protein. In this study, we present evidence that cell-surface expression of IFN-gammaR1 is transiently down-regulated on the surface of naive CD4+ T cells shortly after TCR engagement. Furthermore, this down-regulation is not mediated by the ligand, IFN-gamma, but results from TCR engagement and can be inhibited by cyclosporin A.


Subject(s)
Down-Regulation/immunology , Interferon-gamma/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Interferon/antagonists & inhibitors , Receptors, Interferon/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Extracellular Space/immunology , Extracellular Space/metabolism , Female , Interferon-gamma/genetics , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Ligands , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/physiology , Receptors, Interferon/biosynthesis , Receptors, Interferon/genetics , Signal Transduction/immunology , Time Factors , Interferon gamma Receptor
16.
J Exp Med ; 191(2): 213-24, 2000 Jan 17.
Article in English | MEDLINE | ID: mdl-10637267

ABSTRACT

Cellular interleukin 10s (cIL-10s) of human and murine origin have extensive sequence and structural homology to the Epstein-Barr virus BCRF-I gene product, known as viral IL-10 (vIL-10). Although these cytokines share many immunosuppressive properties, vIL-10 lacks several of the immunostimulatory activities of cIL-10 on certain cell types. The molecular and cellular bases for this dichotomy are not currently defined. Here, we show that the single amino acid isoleucine at position 87 of cIL-10 is required for its immunostimulatory function. Substitution of isoleucine in cIL-10 with alanine, which corresponds to the vIL-10 residue, abrogates immunostimulatory activity for thymocytes, mast cells, and alloantigenic responses while preserving immunosuppressive activity for inhibition of interferon gamma production and prolongation of cardiac allograft survival. Conversely, substitution of alanine with isoleucine in vIL-10 converts it to a cIL-10-like molecule with immunostimulatory activity. This single conservative residue alteration significantly affects ligand affinity for receptor; however, affinity changes do not necessarily alter specific activities for biologic responses in a predictable fashion. These results suggest complex regulation of IL-10 receptor-ligand interactions and subsequent biological responses. These results demonstrate that vIL-10 may represent a captured and selectively mutated cIL-10 gene that benefits viral pathogenesis by leading to ineffective host immune responses. The ability to manipulate the activity of IL-10 in either a stimulatory or suppressive direction may be of practical value for regulating immune responses for disease therapy, and of theoretical value for determining what aspects of IL-10 activity are important for normal T cell responses.


Subject(s)
Interleukin-10/immunology , Isoleucine/immunology , Milk Proteins , Alanine/immunology , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , CHO Cells , COS Cells , Cell Division , Cells, Cultured , Cricetinae , DNA-Binding Proteins/metabolism , Humans , Immune Tolerance , Interleukin-10/chemistry , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Molecular Sequence Data , Receptors, Interleukin/immunology , Receptors, Interleukin/metabolism , Receptors, Interleukin-10 , STAT1 Transcription Factor , STAT3 Transcription Factor , STAT5 Transcription Factor , Sequence Homology, Amino Acid , Trans-Activators/metabolism
17.
Genes Immun ; 1(7): 442-50, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11196675

ABSTRACT

Interleukin-10 (IL-10) is a pleiotropic cytokine with important immunoregulatory functions whose actions influence activities of many of the cell-types in the immune system. We report here identification and cloning of a gene and corresponding cDNAs encoding a novel homologue of IL-10, designated IL-19. IL-19 shares 21% amino acid identity with IL-10. The exon/intron structure of IL-19 is similar to that of the human IL-10 gene, comprising five exons and four introns within the coding region of the IL-19 cDNA. There are at least two distinct IL-19 mRNA species that differ in their 5'-sequences, suggesting the existence of an intron in the 5'-sequences of coding portion of the IL-19 gene. The longer 5'-sequence contains an alternative initiating ATG codon that is in-frame with the rest of the coding sequence. The expression of IL-19 mRNA can be induced in monocytes by LPS-treatment. The appearance of IL-19 mRNA in LPS-stimulated monocytes was slightly delayed compared to expression of IL-10 mRNA: significant levels of IL-10 mRNA were detectable at 2 h post-stimulation, whereas IL-19 mRNA was not detectable until 4 h. Treatment of monocytes with IL-4 or IL-13 did not induce de novo expression of IL-19, but these cytokines did potentiate IL-19 gene expression in LPS-stimulated monocytes. In addition, GM-CSF was capable of directly inducing IL-19 gene expression in monocytes. IL-19 does not bind or signal through the canonical IL-10 receptor complex, suggesting existence of an IL-19 specific receptor complex, the identity of which remains to be discovered.


Subject(s)
Interleukin-10/genetics , 5' Untranslated Regions , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA Primers/genetics , DNA, Complementary/genetics , Humans , Interleukins , Molecular Sequence Data , Monocytes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid
18.
Biopolymers ; 55(4): 254-87, 2000.
Article in English | MEDLINE | ID: mdl-11169919

ABSTRACT

Interferon (IFN) was approved by the U.S. Food and Drug Administration on June 5, 1986. As the first biotherapeutic approved, IFN-alpha paved the way for development of many other cytokines and growth factors. Nevertheless, we have just touched the surface of understanding the multitude of human IFNs. This paper reviews the history of the purification of human leukocyte IFN and key aspects of our current state of knowledge of human interferon alpha genes, proteins, and receptors.


Subject(s)
Interferon-alpha/isolation & purification , Receptors, Interferon/isolation & purification , Animals , Chromatography, High Pressure Liquid , Humans , Interferon Type I/genetics , Interferon Type I/isolation & purification , Interferon-alpha/genetics , Interferon-alpha/pharmacology , Models, Biological , Receptor, Interferon alpha-beta , Receptors, Interferon/genetics , Recombinant Proteins , Sequence Analysis, Protein , Signal Transduction , Interferon gamma Receptor
19.
J Infect Dis ; 181(1): 379-84, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10608793

ABSTRACT

Complete deficiency of either of the two human interferon (IFN)-gamma receptor components, the ligand-binding IFN-gammaR1 chain and the signaling IFN-gammaR2 chain, is invariably associated with early-onset infection caused by bacille Calmette-Guérin vaccines and/or environmental nontuberculous mycobacteria, poor granuloma formation, and a fatal outcome in childhood. Partial IFN-gammaR1 deficiency is associated with a milder histopathologic and clinical phenotype. Cells from a 20-year-old healthy person with a history of curable infections due to bacille Calmette-Guérin and Mycobacterium abscessus and mature granulomas in childhood were investigated. There was a homozygous nucleotide substitution in IFNGR2, causing an amino acid substitution in the extracellular region of the encoded receptor. Cell surface IFN-gammaR2 were detected by flow cytometry. Cellular responses to IFN-gamma were impaired but not abolished. Transfection with the wild-type IFNGR2 gene restored full responsiveness to IFN-gamma. This is the first demonstration of partial IFN-gammaR2 deficiency in humans.


Subject(s)
BCG Vaccine/adverse effects , Mycobacterium Infections/immunology , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Adult , DNA-Binding Proteins/metabolism , Female , Genotype , HLA-DR Antigens/metabolism , Homozygote , Humans , Mycobacterium bovis/immunology , Mycobacterium bovis/pathogenicity , Pedigree , Phenotype , Point Mutation , STAT1 Transcription Factor , Signal Transduction , Trans-Activators/metabolism , Interferon gamma Receptor
20.
Protein Expr Purif ; 17(2): 203-14, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10545268

ABSTRACT

Labeled proteins are used in a variety of applications. This review focuses on methods that utilize genetic engineering to introduce protein kinase recognition sites into proteins. Many protein kinase recognition sites can be introduced into proteins and serve as useful tags for a variety of purposes. The introduction of protein kinase recognition sites into proteins can be achieved without modifying the essential structure or function of the proteins. Because proteins modified by these procedures retain their activity after phosphorylation, they can be used in many applications. The phosphorylatable proteins can be labeled easily to high specific activity with radioisotopes ((32)P, (33)P, or (35)S), or the nonradioactive (31)P can be used. The use of these radioisotopes provides a convenient and safe method for radiolabeling proteins. Moreover, the use of the nonradioactive (31)P with protein tyrosine kinase recognition sites permits the tagging of proteins and their detection with the many anti-phosphotyrosine antibodies available. Overall, the procedure represents a convenient, safe, and efficient method to label proteins for a variety of applications.


Subject(s)
Binding Sites/genetics , Protein Engineering/methods , Protein Kinases/metabolism , Recombinant Proteins/genetics , Amino Acid Motifs/genetics , Animals , Humans , Isotope Labeling/methods , Phosphorylation , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL