Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters










Publication year range
1.
Oncogene ; 35(4): 448-58, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-25867061

ABSTRACT

MicroRNAs (miRNAs) have been demonstrated to have critical roles in regulating cancer cell proliferation, survival and sensitivity to chemotherapy. The potential application of using miRNAs to predict therapeutic response to cancer treatment holds high promise, but miRNAs with predictive value remain to be identified and underlying mechanisms have not been completely understood. Here, we show a strong correlation between miR-621 expression and chemosensitivity to paclitaxel plus carboplatin (PTX/CBP) regimen, an effective neoadjuvant chemotherapy for breast cancer patients. High level of miR-621 predicts better response to PTX/CBP regimen neoadjuvant chemotherapy in breast cancer patients, who also tend to achieve pathological complete response. Ectopic overexpression of miR-621 promoted apoptosis and increased chemosensitivity to PTX and CBP both in cultured breast cancer cells and in xenograft tumor model. We further show that FBXO11 is a direct functional target of miR-621 and miR-621 level is negatively correlated with FBXO11 expression in breast cancer patients. Ectopic expression of FBXO11 attenuated increased apoptosis in breast cancer cells overexpressing miR-621 upon PTX or CBP treatment. Consistently, high FBXO11 expression significantly correlated with poor survival in breast cancer patients. Mechanistically, we found in breast cancer cells FBXO11 interacts with p53 and promotes its neddylation, which suppressed the p53 transactivity. Accordingly, miR-621-dependent FBXO11 suppression enhanced p53 activity and increased apoptosis in breast cancer cells exposed to chemotherapeutics. Taken together, our data suggest that miR-621 enhances chemosensitivity of breast cancer cells to PTX/CBP chemotherapy by suppressing FBXO11-dependent inhibition of p53. miR-621 may serve as a predictive biomarker and a potential therapeutic target in breast cancer treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , F-Box Proteins/genetics , MicroRNAs/genetics , Protein-Arginine N-Methyltransferases/genetics , Tumor Suppressor Protein p53/genetics , Adult , Animals , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carboplatin/administration & dosage , Cell Line, Tumor/drug effects , F-Box Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, Inbred BALB C , MicroRNAs/metabolism , Middle Aged , Paclitaxel/administration & dosage , Protein-Arginine N-Methyltransferases/metabolism , Treatment Outcome , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
2.
Br J Cancer ; 105(12): 1874-84, 2011 Dec 06.
Article in English | MEDLINE | ID: mdl-22095230

ABSTRACT

BACKGROUND: Melanoma is highly resistant to current modalities of therapy, with the extent of pigmentation playing an important role in therapeutic resistance. Nuclear factor-κB (NF-κB) is constitutively activated in melanoma and can serve as a molecular target for cancer therapy and steroid/secosteroid action. METHODS: Cultured melanoma cells were used for mechanistic studies on NF-κB activity, utilising immunofluorescence, western blotting, EMSA, ELISA, gene reporter, and estimated DNA synthesis assays. Formalin-fixed, paraffin-embedded specimens from melanoma patients were used for immunocytochemical analysis of NF-κB activity in situ. RESULTS: Novel 20-hydroxyvitamin (20(OH)D(3)) and classical 1α,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) secosteroids inhibited melanoma cell proliferation. Active forms of vitamin D were found to inhibit NF-κB activity in nonpigmented cells, while having no effect on pigmented cells. Treatment of nonpigmented cells with vitamin D3 derivatives inhibited NF-κB DNA binding and NF-κB-dependent reporter assays, as well as inhibited the nuclear translocation of the p65 NF-κB subunit and its accumulation in the cytoplasm. Moreover, analysis of biopsies of melanoma patients showed that nonpigmented and slightly pigmented melanomas displayed higher nuclear NF-κB p65 expression than highly pigmented melanomas. CONCLUSION: Classical 1,25(OH)(2)D(3) and novel 20(OH)D(3) hydroxyderivatives of vitamin D3 can target NF-κB and regulate melanoma progression in nonpigmented melanoma cells. Melanin pigmentation is associated with the resistance of melanomas to 20(OH)D(3) and 1,25(OH)(2)D(3) treatment.


Subject(s)
Cholecalciferol/pharmacology , Melanoma/metabolism , NF-kappa B/metabolism , Cell Division/drug effects , Cell Line, Tumor , Humans , Melanoma/pathology
3.
Leukemia ; 24(4): 806-12, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20130599

ABSTRACT

Acute lymphoblastic leukemia (ALL) harboring the t(4;11) translocation is associated with a very poor prognosis; innovative treatment strategies are required to improve the current 5-year survival rate of 30-40%. Interferon beta (IFN beta) has shown promise in the treatment of both solid and hematologic malignancies, although the short half-life and toxicity associated with high doses have limited its clinical utility. To overcome these limitations, we investigated the effect of continuous, gene transfer-mediated delivery of IFN beta using adeno-associated virus (AAV)-mediated expression, on ALL cells with the t(4;11) translocation. We found that this method of IFN beta delivery resulted in complete remission of leukemia in a murine model. However, leukemic cells eventually became resistant to IFN beta and relapse was observed. Activation of NF-kappaB was identified as a mechanism for IFN beta resistance, and inhibition of NF-kappaB activity in resistant cells sensitized cells to IFN beta. IFN beta combined with agents that inhibit NF-kappaB could have therapeutic potential in the treatment of children with mixed lineage leukemia subtype ALL.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Leukemic , Gene Rearrangement , Interferon-beta/pharmacology , Myeloid-Lymphoid Leukemia Protein/genetics , NF-kappa B/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 4/genetics , Dependovirus/genetics , Drug Resistance, Neoplasm , Electrophoretic Mobility Shift Assay , Flow Cytometry , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, SCID , Myeloid-Lymphoid Leukemia Protein/metabolism , NF-kappa B/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Translocation, Genetic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
J Biol Chem ; 276(49): 45909-13, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11590175

ABSTRACT

The proliferation of the rat intestinal mucosal IEC-6 cell line requires polyamines, whose synthesis is catalyzed by the enzyme ornithine decarboxylase (ODC). ODC inhibition leads to polyamine depletion, as well as inhibition of both cell proliferation and apoptosis by regulating gene expression. The NF-kappa B transcription factor regulates genes involved in apoptotic, immune, and inflammatory responses. In the present study we tested the hypothesis that NF-kappa B is activated following ODC inhibition. We found that the inhibition of ODC by alpha-difluoromethylornithine (DFMO) resulted in a approximately 50% decrease in intracellular putrescine levels within 1 h. NF-kappa B is activated by DFMO through the degradation of the inhibitory protein I kappa B alpha that sequesters NF-kappa B in the cytoplasm. The DFMO-induced NF-kappa B complexes contain the p65 and p50 members of the Rel protein family. DFMO-induced NF-kappa B activation was accompanied by the translocation of p65 from the cytoplasm into the nucleus. DFMO selectively inhibited a gene reporter construct dependent on the kappa B site present in the HLA-B7 gene. In contrast, DFMO had no effect on a gene reporter construct dependent on the kappa B site present in the interleukin-8 gene. Thus, we report that ODC inhibition activates the NF-kappa B transcription factor, which may mediate the altered physiological state of intestinal cells that occurs following polyamine depletion.


Subject(s)
Biogenic Polyamines/physiology , I-kappa B Proteins , NF-kappa B/metabolism , Animals , Biogenic Polyamines/metabolism , Cell Line , DNA-Binding Proteins/metabolism , Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , Hydrolysis , Immunohistochemistry , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , NF-KappaB Inhibitor alpha , Ornithine Decarboxylase/metabolism , Ornithine Decarboxylase Inhibitors , Rats
6.
J Biol Chem ; 276(17): 13756-61, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11278812

ABSTRACT

Interferons (IFNs) play critical roles in host defense by modulating gene expression via activation of signal transducer and activator of transcription (STAT) factors. IFN-alpha/beta also activates another transcription factor, nuclear factor kappaB (NF-kappaB), which protects cells against apoptotic stimuli. NF-kappaB activation requires the IFN-dependent association of STAT3 with the IFNAR1 chain of the IFN receptor. IFN-dependent NF-kappaB activation involves the sequential activation of a serine kinase cascade involving phosphatidylinositol 3-kinase (PI-3K) and Akt. Whereas constitutively active PI-3K and Akt induce NF-kappaB activation, Ly294002 (a PI-3K inhibitor), dominant-negative PI-3K, and kinase-dead Akt block IFN-dependent NF-kappaB activation. Moreover, dominant-negative PI-3K blocks IFN-promoted degradation of kappaBox alpha. Ly294002, a dominant-negative PI-3K construct, and kinase-dead Akt block IFN-promoted cell survival, enhancing apoptotic cell death. Therefore, STAT3, PI-3K, and Akt are components of an IFN signaling pathway that promotes cell survival through NF-kappaB activation.


Subject(s)
Interferon-alpha/physiology , Interferon-beta/physiology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Apoptosis , Cell Death , Cell Nucleus/metabolism , Cell Survival , Chromones/pharmacology , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Genes, Dominant , Humans , Morpholines/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt , Recombinant Proteins/metabolism , STAT3 Transcription Factor , Signal Transduction , Time Factors , Trans-Activators/metabolism , Transfection
7.
Proc Natl Acad Sci U S A ; 97(25): 13631-6, 2000 Dec 05.
Article in English | MEDLINE | ID: mdl-11095741

ABSTRACT

IFNs play critical roles in host defense by modulating the expression of various genes via signal transducer and activator of transcription factors. We show that IFNalpha/beta activates another important transcription factor, NF-kappaB. DNA-binding activity of NF-kappaB was induced by multiple type 1 IFNs and was promoted by IFN in a diverse group of human, monkey, rat, and murine cells. Human IFN promoted NF-kappaB activation in murine cells that express the human IFNalpha/beta receptor-1 signal-transducing chain of the type 1 IFN receptor. IFN promotes inhibitor of kappa B alpha (IkappaBalpha) serine phosphorylation and degradation, and stimulates NF-kappaB DNA-binding and transcriptional activity. Importantly, IFN promotes cell survival by protecting cells against a variety of proapoptotic stimuli, such as virus infection and antibody-mediated crosslinking. Expression of superrepressor forms of IkappaBalpha, besides inhibiting IFN-mediated NF-kappaB activation and IkappaBalpha degradation, also enhanced apoptotic cell death in IFN-treated cells. We conclude that NF-kappaB activation by IFNalpha/beta is integrated into a signaling pathway through the IFNalpha/beta receptor-1 chain of the type 1 IFN receptor that promotes cell survival in apposition to various apoptotic stimuli.


Subject(s)
Cell Survival/physiology , Interferon-alpha/physiology , Interferon-beta/physiology , NF-kappa B/metabolism , Apoptosis/physiology , Base Sequence , DNA Probes , Hydrolysis , I-kappa B Proteins/chemistry , I-kappa B Proteins/metabolism , Phosphorylation , Serine/metabolism
8.
J Interferon Cytokine Res ; 20(9): 787-94, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11032398

ABSTRACT

Retinoic acid (RA) can potentiate the antitumor effect of interferons (IFN) in a variety of tumor types, including renal cell carcinoma (RCC). The mechanisms by which RA and IFN increase the antitumor effects in RCC are unknown. We used growth assays and mobility shift assays to examine the effects of combining 13-cis-retinoic acid (CRA) and IFN-alpha (plus IFN-gamma) on proliferation and on the expression of the IFN-specific transcription factor IFN-stimulated gene factor 3 (ISGF3) in RCC cell lines. Combining CRA and IFN-alpha resulted in a significant increase in growth inhibition in four cell lines compared with IFN-alpha or CRA alone. Binding of nuclear extracts from RCC cells to an IFN-stimulated response element (ISRE) oligonucleotide probe following incubation with IFN-alpha was not increased by CRA but was significantly increased by pretreatment by IFN-gamma in a time-dependent fashion. Proliferation assays showed that sequential addition of IFN-gamma and IFN-alpha significantly increased growth inhibition. IFN-alpha but not IFN-gamma or CRA increased the cellular levels Stat2 and p48 but not Statl. IFN-gamma pretreatment enhanced the upregulation of p48 levels by IFN-alpha. Combining RA and IFN results in additive growth inhibition on RCC cell lines. This increase in growth inhibition is not mediated by increased ISGF3 expression.


Subject(s)
Antineoplastic Agents/pharmacology , Interferon-alpha/pharmacology , Tretinoin/pharmacology , Carcinoma, Renal Cell/pathology , Cell Division/drug effects , Drug Interactions , Drug Synergism , Humans , Interferon-gamma/pharmacology , Isotretinoin/pharmacology , Kidney Neoplasms/pathology , Signal Transduction/drug effects , Tumor Cells, Cultured
10.
Am J Physiol Cell Physiol ; 278(2): C331-5, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10666028

ABSTRACT

Polyamines are required for the proliferation of the rat intestinal mucosal IEC-6 cell line. Ornithine decarboxylase (ODC) is the enzyme that catalyzes the first step in polyamine synthesis. ODC inhibition not only leads to polyamine depletion but also leads to inhibition of cell proliferation and regulates the expression of the immediate-early genes c-fos, c-myc, and c-jun. Members of the signal transducers and activators of transcription (STAT) transcription factor family bind to the sis-inducible element (SIE) present in the promoters to regulate the expression of a variety of important genes. In the present study, we tested the hypothesis that the STAT3 transcription factor, which is responsible for activation of the acute phase response genes, is activated after inhibition of ODC. We found that inhibition of ODC rapidly induces STAT3 activation as determined by STAT3 tyrosine phosphorylation, translocation of STAT3 from the cytoplasm into the nucleus, and the presence of STAT3 in SIE-dependent DNA-protein complexes. STAT3 activation upon inhibition of ODC was accompanied by the activation of a STAT3-dependent reporter construct. Moreover, prolonged polyamine depletion resulted in downregulation of cellular STAT3 levels.


Subject(s)
DNA-Binding Proteins/metabolism , Epithelial Cells/enzymology , Intestines/cytology , Ornithine Decarboxylase Inhibitors , Trans-Activators/metabolism , Tyrosine/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Choline O-Acetyltransferase/genetics , Cytoplasm/metabolism , DNA Probes , Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Gene Expression Regulation, Enzymologic , Genes, Reporter , Phosphorylation , Polyamines/metabolism , Promoter Regions, Genetic/physiology , Rats , STAT3 Transcription Factor , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptional Activation/drug effects
11.
Nature ; 401(6748): 82-5, 1999 Sep 02.
Article in English | MEDLINE | ID: mdl-10485710

ABSTRACT

Activation of the nuclear transcription factor NF-kappaB by inflammatory cytokines requires the successive action of NF-kappaB-inducing kinase (NIK) and an IKB-kinase (IKK) complex composed of IKKalpha and IKKbeta. Here we show that the Akt serine-threonine kinase is involved in the activation of NF-kappaB by tumour necrosis factor (TNF). TNF activates phosphatidylinositol-3-OH kinase (PI(3)K) and its downstream target Akt (protein kinase B). Wortmannin (a PI(3)K inhibitor), dominant-negative PI(3)K or kinase-dead Akt inhibits TNF-mediated NF-kappaB activation. Constitutively active Akt induces NF-kappaB activity and this effect is blocked by dominant-negative NIK. Conversely, NIK activates NF-kappaB and this is blocked by kinase-dead Akt. Thus, both Akt and NIK are necessary for TNF activation of NF-kappaB. Akt mediates IKKalpha phosphorylation at threonine 23. Mutation of this amino acid blocks phosphorylation by Akt or TNF and activation of NF-kappaB. These findings indicate that Akt is part of a signalling pathway that is necessary for inducing key immune and inflammatory responses.


Subject(s)
NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Antigens, CD/metabolism , Cell Line , DNA/metabolism , Enzyme Activation , HeLa Cells , Humans , I-kappa B Kinase , Mice , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I , Signal Transduction , Threonine/metabolism , NF-kappaB-Inducing Kinase
12.
Am J Physiol ; 276(2): C419-25, 1999 02.
Article in English | MEDLINE | ID: mdl-9950769

ABSTRACT

Signal transducers and activators of transcription (STATs) are cytoplasmic proteins that bind to activated membrane receptors, undergo ligand-dependent phosphorylation on tyrosine residues, and translocate to the nucleus, where they induce transcription of specific genes in response to a variety of ligands, including cytokines and some growth factors. Using immunocytochemical and biochemical techniques, we investigated the localization and responses of STAT1 and STAT2 to epidermal growth factor (EGF) stimulation in IEC-6 intestinal epithelial cells and HeLa cells. These studies provide the first description of STAT activation and localization in response to EGF in intestinal epithelial cells and some novel findings regarding the activation and localization of STATs in general. These include the following. First, EGF promoted the tyrosine phosphorylation of STAT1 in IEC-6 cells and caused its translocation to the nucleus. Second, in the absence of EGF stimulation both STAT1 and STAT2 were localized to the Golgi apparatus in IEC-6 cells. Third, EGF caused the translocation of STAT2 to the nucleus in both IEC-6 and HeLa cells without inducing the tyrosine phosphorylation of STAT2.


Subject(s)
DNA-Binding Proteins/metabolism , Epidermal Growth Factor/pharmacology , Intestinal Mucosa/metabolism , Trans-Activators/metabolism , Tyrosine/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , HeLa Cells , Humans , Interferons/pharmacology , Intestinal Mucosa/cytology , Phosphorylation/drug effects , STAT1 Transcription Factor , STAT2 Transcription Factor , Tissue Distribution/drug effects
13.
Cancer Res ; 58(12): 2489-99, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9635566

ABSTRACT

IFNs were first described as potent antiviral agents 40 years ago, and recombinant IFN-alpha2a and IFN-alpha2b were approved for the treatment of hairy cell leukemia just 11 years ago. Today, alpha-IFNs are approved worldwide for the treatment of a variety of malignancies and virologic diseases. Although the exact mechanism of action of IFN-alpha in the treatment of such diseases is not fully understood, many advances have been made in the characterization of the physicochemical and diverse biological properties of this highly pleiotropic cytokine. Here we review recent developments in our understanding of the antiviral and immunoregulatory properties of IFN-alpha, the nature of the multisubunit IFN-alpha receptor, and the molecular mechanisms of signal transduction. Where available, we have included comparative data on recombinant alpha-IFNs derived from both naturally occurring and nonnaturally occurring synthetic genes. We also review clinical data and data on the side effects and antigenicity of different sources of recombinant alpha-IFNs in humans. These latter topics are of clinical interest, because they may potentially affect the efficacy of these various products. Hopefully, what is already known about IFN will prompt further exploration into the mechanism(s) of action of IFN-alpha and thus deliver new applications for this prototypic cytokine, whose full therapeutic potential is yet to be realized.


Subject(s)
Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Interferon Type I/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Antiviral Agents/therapeutic use , Binding, Competitive , Cytokines/antagonists & inhibitors , Dinoprostone/physiology , Forecasting , Humans , Interferon Type I/chemistry , Interferon Type I/metabolism , Interferon Type I/therapeutic use , Neoplasms/drug therapy , Protein Conformation , Receptor, Interferon alpha-beta , Receptors, Interferon/drug effects , Receptors, Interferon/metabolism , Recombinant Proteins , Signal Transduction , Treatment Outcome , Virus Diseases/drug therapy
14.
Proc Natl Acad Sci U S A ; 95(10): 5568-72, 1998 May 12.
Article in English | MEDLINE | ID: mdl-9576923

ABSTRACT

STAT proteins play critical roles in the signal transduction pathways for various cytokines. The type I interferons (IFNalpha/beta) promote the DNA-binding activity of the transcription factors STAT1, STAT2, and STAT3. Although the requirement for STAT1 and STAT2 in IFNalpha/beta signaling and action is well documented, the biological importance of STAT3 to IFN action has not yet been addressed. We found that STAT3 plays a critical role in signal transduction by IFNalpha/beta. A human cell line that is resistant to the antiviral and antiproliferative activities of IFN but is still IFN-responsive by virtue of STAT1 and STAT2 activation was found to be defective in STAT3 activation and in induction of NF-kappaB DNA-binding activity. Expression of STAT3 in these resistant cells complemented these signaling defects and also markedly increased cellular sensitivity to the antiviral and antiproliferative effects of IFN. Because STAT3 is involved in the induction of NF-kappaB DNA-binding activity and in the induction of antiviral and antiproliferative activity, our results place STAT3 as an important upstream element in type I IFN signal transduction and in the induction of biological activities. Therefore, our results indicate that STAT1 and STAT2 are not the only STATs required for the expression of the key biological activities of IFNalpha/beta.


Subject(s)
DNA-Binding Proteins/metabolism , Interferon Type I/physiology , Signal Transduction , Trans-Activators/metabolism , Antiviral Agents/metabolism , Cell Division/drug effects , Drug Resistance , Humans , NF-kappa B/metabolism , Phosphorylation , Ribosomal Protein S6 Kinases/metabolism , STAT2 Transcription Factor , STAT3 Transcription Factor , Tumor Cells, Cultured , Tyrosine/metabolism
15.
Virology ; 242(1): 14-21, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9501047

ABSTRACT

The first cloned chain (IFNAR1) of the human interferon-alpha (IFN alpha) receptor acts as a species-specific transducer for type 1 IFN action when transfected into heterologous mouse cells. Stably transfected mouse L929 cell lines expressing truncation mutants of the intracellular domain of the human IFNAR1 chain were tested for biological responses to human IFN alpha. Deletion of the intracellular domain resulted in a complete loss of sensitivity to the biological activity of human IFN but markedly increased IFNAR1 cell surface expression, demonstrating that the intracellular domain is required for biological function and contains a domain that negatively regulates its cell surface expression. Removal of the conserved membrane distal 16-amino-acid IRTAM (Interferon Receptor Tyrosine Activation Motif) sequence: (1) increased sensitivity to IFN alpha's antiviral activity, (2) increased the rapid IFN alpha-dependent formation of STAT-containing DNA-binding complexes, (3) prolonged tyrosine phosphorylation kinetics of the JAK-STAT pathway, and (4) blocked the IFN-dependent down-regulation of the IFNAR1 chain. These results indicate that the IRTAM negatively regulates signalling events required for the induction of IFN's biological actions via regulating receptor down-regulation.


Subject(s)
Antiviral Agents/pharmacology , DNA-Binding Proteins/metabolism , Interferon Type I/pharmacology , Protein-Tyrosine Kinases , Proteins/metabolism , Receptors, Interferon/physiology , Trans-Activators/metabolism , Amino Acid Sequence , Animals , Cell Line , Conserved Sequence , Humans , L Cells , Membrane Proteins , Mice , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Interferon alpha-beta , Receptors, Interferon/biosynthesis , Receptors, Interferon/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , STAT2 Transcription Factor , Sequence Deletion , Signal Transduction , TYK2 Kinase , Transfection
16.
J Immunol ; 160(6): 2742-50, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9510175

ABSTRACT

Cellular responses to TNF are initiated by either of two cell surface receptors, the type 1 TNF receptor (TNFR1) and the type 2 TNF receptor (TNFR2). Although neither receptor contains an intrinsic protein tyrosine kinase, such activity has been implicated in TNF action. In this study, we show that murine TNF induces the tyrosine phosphorylation and activation of the intracellular Janus tyrosine kinases Jak1, Jak2, and Tyk2 in murine 3T3-L1 adipocytes. Activation of Jak kinases by TNF was associated with tyrosine phosphorylation of STAT1, STAT3, STAT5, and STAT6, but not STAT2 or STAT4, showing that TNF acts on a specific subset of these latent cytoplasmic transcription factors in 3T3-L1 adipocytes. Agonist antiserum to TNFR1 induced Jak kinase and STAT protein phosphorylation. Phosphorylation of Jak proteins was also induced by human TNF, which selectively binds to TNFR1 on murine cells. 35S-labeled Jak kinases were precipitated from a cell-free system and from lysates of 3T3-L1 adipocytes by a glutathione S-transferase fusion protein containing the cytoplasmic domain of TNFR1. These results suggest that the cytoplasmic domain of TNFR1 can directly interact with and form signaling complexes with Jak kinases. Jak2 was precipitated from HeLa cells by antiserum to TNFR1, directly demonstrating their association in vivo. Thus, TNF activates a Jak/STAT signal-transduction cascade by acting through TNFR1.


Subject(s)
DNA-Binding Proteins/physiology , Milk Proteins , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins , Receptors, Tumor Necrosis Factor/physiology , Trans-Activators/physiology , 3T3 Cells , Animals , DNA/metabolism , Humans , Janus Kinase 1 , Janus Kinase 2 , Mice , Phosphorylation , Rabbits , STAT1 Transcription Factor , STAT3 Transcription Factor , STAT5 Transcription Factor , STAT6 Transcription Factor , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , Tyrosine/metabolism
17.
Cancer Biother Radiopharm ; 13(3): 143-54, 1998 Jun.
Article in English | MEDLINE | ID: mdl-10850350

ABSTRACT

Recombinant alfa interferons (IFN-alpha s) are approved worldwide for the treatment of a variety of cancers and diseases of virologic origin. A series of recent advances in the molecular characterization of recombinant IFN-alpha s have allowed the determination of the three-dimensional IFN-alpha 2b structure by high-resolution x-ray crystallography. We review here recent developments in our understanding of the molecular and physicochemical properties of recombinant IFN-alpha, including our current state of knowledge of the IFN-alpha gene family and the multiple species of human leukocyte IFN. Based on the reported three-dimensional structure of IFN-alpha 2b, we propose a molecular model for the IFN-alpha 2b receptor complex and predict models for the naturally occurring subtypes IFN-alpha 1 and IFN-alpha 8, as well as the synthetic, non-naturally occurring consensus IFN. Such models provide molecular insights into the mechanism of action of IFN-alpha.


Subject(s)
Interferon Type I/chemistry , Interferon Type I/therapeutic use , Neoplasms/drug therapy , Virus Diseases/drug therapy , Humans , Interferon alpha-2 , Interferon-alpha/chemistry , Interferon-alpha/genetics , Models, Molecular , Protein Conformation , Receptors, Interferon/chemistry , Recombinant Proteins
18.
J Biol Chem ; 272(34): 21045-51, 1997 Aug 22.
Article in English | MEDLINE | ID: mdl-9261106

ABSTRACT

The type I interferons (IFNs) are a family of cytokines, comprising at least 17 subtypes, which exert pleiotropic actions by interaction with a multi-component cell surface receptor and at least one well characterized signal transduction pathway involving JAK/STAT (Janus kinase/signal transducer and activator of transcription) proteins. In a previous report, we showed that a signaling factor, encoded by a gene located on the distal portion of chromosome 21, distinct from the IFNAR-1 receptor, was necessary for 2'-5'-oligoadenylate synthetase activity and antiviral responses, but not for high affinity ligand binding. In the present studies using hybrid Chinese hamster ovary cell lines containing portions of human chromosome 21, we show that the type I IFN signaling molecule, designated herein as ISF21, is distinct from the second receptor component, IFNAR-2, which is expressed in signaling and non-signaling cell lines. The location of the gene encoding ISF21 is narrowed to a region between the 10;21 and the r21 breakpoints, importantly eliminating the Mx gene located at 21q22.3 (the product of which is involved in IFN-induced antiviral responses) as a candidate for the signaling factor. To characterize the action of this factor in the type I IFN signaling pathway, we show that it acts independently of receptor down-regulation following ligand binding, both of which occur equally in the presence or absence of the factor. In addition, we demonstrate that ISF21 is necessary for transcriptional activation of 2'-5'-oligoadenylate synthetase, 6-16, and guanylate-binding protein gene promoter reporter constructs, which are mediated by several signaling pathways. ISF21 represents a novel factor as the localization to chromosome 21, and the data presented in this study exclude any of the known type I IFN signal-transducing molecules.


Subject(s)
Biological Factors/physiology , Interferon Type I/physiology , Receptors, Interferon/physiology , Animals , CHO Cells , Chromosome Mapping , Chromosomes, Human, Pair 21 , Cricetinae , Down-Regulation , Humans , Hybrid Cells , Membrane Proteins , RNA, Messenger/genetics , Receptor, Interferon alpha-beta , Signal Transduction , Transcriptional Activation
19.
Semin Oncol ; 24(3 Suppl 9): S9-63-S9-69, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9208874

ABSTRACT

Because alpha-interferon (IFN-alpha) has a number of therapeutic applications in the treatment of various human cancers and diseases of viral origin, an understanding of how this family of proteins interacts with cells to induce their pleiotropic biologic activities is essential. Available data suggest that recombinant IFN-alphas from both natural and synthetic genes bind to a common cell surface receptor and induce antiviral activity in a variety of cell lines. IFN-alphas were found to differ significantly in their abilities to bind to cells; this difference varied with the types of IFN-alpha and cell type used. Consensus interferon (IFN-con1), a nonnaturally occurring synthetic IFN, and IFN-alpha2b competed about equally well for receptor binding sites on Daudi and CaKi cells and were followed by IFN-alpha8 in the ability to compete. Results of affinity cross-linking experiments indicated that all three IFN-alphas interacted similarly with the multichain IFN-alpha receptor. IFN-alpha7, however, competed poorly for binding sites on both cell lines. Each of the IFN-alphas tested displayed discrete biologic differences, which also varied with the assay system used. IFN-con1 and IFN-alpha2b displayed similar antiviral activities on CaKi cells using vesicular stomatitis virus; the viral activities of these IFNs were significantly greater than those of IFN-alpha7 or IFN-alpha8. Studies with murine transfectants demonstrated significant differences in the various IFNs to interact with the IFN-alpha receptor-1 chain of the type I IFN receptor. It is yet to be established, however, that these various in vitro distinctions result in differences in clinical benefit or toxicity between the various subtypes.


Subject(s)
Interferon Type I/pharmacology , Interferon-alpha/pharmacology , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Binding, Competitive , Humans , Interferon Type I/metabolism , Interferon-alpha/metabolism , Mice , Receptors, Interferon/metabolism , Recombinant Proteins
20.
Science ; 276(5317): 1418-20, 1997 May 30.
Article in English | MEDLINE | ID: mdl-9162009

ABSTRACT

STAT (signal transducers and activators of transcription) proteins undergo cytokine-dependent phosphorylation on serine and tyrosine. STAT3, a transcription factor for acute phase response genes, was found to act as an adapter molecule in signal transduction from the type I interferon receptor. STAT3 bound to a conserved sequence in the cytoplasmic tail of the IFNAR1 chain of the receptor and underwent interferon-dependent tyrosine phosphorylation. The p85 regulatory subunit of phosphatidylinositol 3-kinase, which activates a series of serine kinases, bound to phosphorylated STAT3 and subsequently underwent tyrosine phosphorylation. Thus, STAT3 acts as an adapter to couple another signaling pathway to the interferon receptor.


Subject(s)
Acute-Phase Proteins/genetics , DNA-Binding Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Interferon/metabolism , Trans-Activators/metabolism , Amino Acid Sequence , Androstadienes/pharmacology , Animals , Binding Sites , COS Cells , Cell Line , Cloning, Molecular , Conserved Sequence , DNA-Binding Proteins/genetics , Enzyme Inhibitors/pharmacology , Membrane Proteins , Molecular Sequence Data , Phosphatidylinositol 3-Kinases , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Point Mutation , Protein Binding , Receptor, Interferon alpha-beta , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , STAT3 Transcription Factor , Signal Transduction , Trans-Activators/genetics , Tyrosine/metabolism , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...