Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Br J Dermatol ; 171(3): 512-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24602010

ABSTRACT

BACKGROUND: Inflammatory chemokines, such as macrophage-derived chemokine (MDC/CCL22), are elevated in the serum and lesioned skin of patients with atopic dermatitis (AD), and are ligands for C-C chemokine receptor 4, which is predominantly expressed on T helper 2 lymphocytes, basophils and natural killer cells. We have previously reported that quercetagetin has an inhibitory activity on inflammatory chemokines, which is induced by interferon (IFN)-γ and tumour necrosis factor (TNF)-α, occurring via inhibition of the signal transducer and activator of transcription 1 (STAT1) signal. OBJECTIVES: To investigate the specific mechanisms of quercetagetin on the STAT1 signal. METHODS: We confirmed the inhibitory activity of quercetagetin on MDC and STAT1 in HaCaT keratinocytes. The interaction between STAT1 and IFN-γR1 was investigated using immunoprecipitation. The small interfering RNA approach was used to investigate the role of suppressor of cytokine signalling 1 (SOCS1) and transforming growth factor (TGF)-ß1 induced by quercetagetin. RESULTS: Quercetagetin inhibited the expression of MDC at both the protein and mRNA levels in IFN-γ- and TNF-α-stimulated HaCaT human keratinocytes. Moreover, quercetagetin inhibited the phosphorylation of STAT1 through upregulation of SOCS1. Increased expression of SOCS1 disrupted the binding of STAT1 to IFN-γR1. Furthermore, quercetagetin augmented the expression of TGF-ß1, which is known to modulate the immune response and inflammation. CONCLUSIONS: These results suggest that quercetagetin may be a potent inhibitor of the STAT1 signal, which could be a new molecular target for anti-inflammatory treatment, and may thus have therapeutic applications as an immune modulator in inflammatory diseases such as AD.


Subject(s)
Chemokine CCL22/antagonists & inhibitors , Chromones/pharmacology , Keratinocytes/drug effects , STAT1 Transcription Factor/drug effects , Transforming Growth Factor beta1/drug effects , Flavones , Humans , Interferon-gamma/drug effects , Janus Kinases/drug effects , Receptors, Interferon/drug effects , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Tumor Necrosis Factor-alpha/drug effects , Interferon gamma Receptor
2.
Clin Transl Med ; 12(2): e716, 2022 02.
Article in English | MEDLINE | ID: mdl-35184395

ABSTRACT

BACKGROUND: Immunotransmitters (e.g., neurotransmitters and neuromodulators) could orchestrate diverse immune responses; however, the elaborated mechanism by which melatonergic activation governs inflammation remains less defined. METHODS: Primary macrophages, various cell lines, and Pasteurella multocida (PmCQ2)-infected mice were respectively used to illustrate the influence of melatonergic signalling on inflammation in vitro and in vivo. A series of methods (e.g., RNA-seq, metabolomics, and genetic manipulation) were conducted to reveal the mechanism whereby melatonergic signalling reduces macrophage inflammation. RESULTS: Here, we demonstrate that melatonergic activation substantially lessens interleukin (IL)-1ß-dependent inflammation. Treatment of macrophages with melatonin rewires metabolic program, as well as remodels signalling pathways which depends on interferon regulatory factor (IRF) 7. Mechanistically, melatonin acts via membrane receptor (MT) 1 to increase heat shock factor (Hsf) 1 expression through lowering the inactive glycogen synthase kinase (GSK3) ß, thereby transcriptionally inhibiting interferon (IFN)-γ receptor (IFNGR) 2 and ultimately causing defective canonical signalling events [Janus kinase (JAK) 1/2-signal transducer and activator of transcription (STAT) 1-IRF7] and lower IL-1ß production in macrophages. Moreover, we find that melatonin amplifies host protective responses to PmCQ2 infection-induced pneumonia. CONCLUSIONS: Our conceptual framework provides potential therapeutic targets to prevent and/or treat inflammatory diseases associating with excessive IL-1ß production.


Subject(s)
Inflammation/drug therapy , Interleukin-1beta/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Receptors, Interferon/drug effects , Animals , China , Disease Models, Animal , Inflammation/physiopathology , Inflammation/prevention & control , Mice , Receptors, Interferon/metabolism , Signal Transduction/drug effects
3.
J Immunol ; 182(11): 7146-54, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19454711

ABSTRACT

Leishmania donovani, a protozoan parasite, resides and replicates as amastigotes within macrophages. The parasite inflicts the disease visceral leishmaniasis by suppressing host cell function. Neither a therapeutic vaccine nor an effective anti-leishmanial drug to reverse the immunosuppression is available. Although miltefosine (hexadecylphosphocholine or HPC) is a promising orally bioavailable anti-leishmanial drug, its efficacy is seriously compromised by contra-indications in pregnant women. Further rational redesigning of the drug requires studies on its mechanism of action, which is unknown at present. Because miltefosine is proposed to have immunomodulatory functions, we examined whether miltefosine exerts its anti-leishmanial functions by activating macrophages. We observed that miltefosine's anti-leishmanial function was significantly compromised in IFN-gamma-deficient macrophages suggesting the importance of endogenous IFN-gamma in miltefosine-induced anti-leishmanial functions of macrophages. Miltefosine induced IFN-gamma, neutralization of which reduced the anti-leishmanial functions of macrophages. IFN-gamma responsiveness is reduced in L. donovani-infected macrophages but is significantly restored by miltefosine, as it enhances IFN-gamma receptors and IFN-gamma induced STAT-1 phosphorylation but reduced activation of SHP-1, the phosphatase implicated in the down-regulation of STAT-1 phosphorylation. Miltefosine induced protein kinase C-dependent and PI3K-dependent p38MAP kinase phosphorylation and anti-leishmanial function. Miltefosine promotes p38MAP kinase-dependent anti-leishmanial functions and IL-12-dependent Th1 response. Leishmania donovani-infected macrophages induced Th2 response but miltefosine treatment reversed the response to Th1-type. Thus, our data define for the first time the mechanistic basis of host cell-dependent anti-leishmanial function of miltefosine.


Subject(s)
Immunity/drug effects , Interferon-gamma/immunology , Phosphorylcholine/analogs & derivatives , Animals , Antiprotozoal Agents/pharmacology , Interferon-gamma/deficiency , Leishmania donovani/immunology , Leishmaniasis, Visceral/drug therapy , Macrophages/parasitology , Mice , Mice, Knockout , Phosphorylation , Phosphorylcholine/pharmacology , Receptors, Interferon/drug effects , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , T-Lymphocytes, Helper-Inducer/drug effects , Interferon gamma Receptor
4.
Mol Cell Biol ; 26(5): 1888-97, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16479007

ABSTRACT

Alpha and beta interferons (IFN-alpha and IFN-beta) are multifunctional cytokines that exhibit differential activities through a common receptor composed of the subunits IFNAR1 and IFNAR2. Here we combined biophysical and functional studies to explore the mechanism that allows the alpha and beta IFNs to act differentially. For this purpose, we have engineered an IFN-alpha2 triple mutant termed the HEQ mutant that mimics the biological properties of IFN-beta. Compared to wild-type (wt) IFN-alpha2, the HEQ mutant confers a 30-fold higher binding affinity towards IFNAR1, comparable to that measured for IFN-beta, resulting in a much higher stability of the ternary complex as measured on model membranes. The HEQ mutant, like IFN-beta, promotes a differentially higher antiproliferative effect than antiviral activity. Both bring on a down-regulation of the IFNAR2 receptor upon induction, confirming an increased ternary complex stability of the plasma membrane. Oligonucleotide microarray experiments showed similar gene transcription profiles induced by the HEQ mutant and IFN-beta and higher levels of gene induction or repression than those for wt IFN-alpha2. Thus, we show that the differential activities of IFN-beta are directly related to the binding affinity for IFNAR1. Conservation of the residues mutated in the HEQ mutant within IFN-alpha subtypes suggests that IFN-alpha has evolved to bind IFNAR1 weakly, apparently to sustain differential levels of biological activities compared to those induced by IFN-beta.


Subject(s)
Interferon-alpha/genetics , Interferon-alpha/pharmacology , Interferon-beta/metabolism , Interferon-beta/pharmacology , Membrane Proteins/metabolism , Receptors, Interferon/metabolism , Antiviral Agents/pharmacology , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation , Gene Expression Regulation , Humans , Interferon-alpha/metabolism , Membrane Proteins/drug effects , Membrane Proteins/genetics , Multiprotein Complexes , Mutation , Promoter Regions, Genetic , Receptor, Interferon alpha-beta , Receptors, Interferon/drug effects , Receptors, Interferon/genetics , Transcriptional Activation
5.
Neuroscience ; 152(3): 785-97, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18304744

ABSTRACT

We determined the nervous system targeting of interferon-beta1b (IFN-beta1b), a 20 kDa protein used to treat the relapsing-remitting form of multiple sclerosis, following intranasal administration in anesthetized, adult cynomolgus monkeys. Five animals received an intranasal bolus of [(125)I]-labeled IFN-beta1b, applied bilaterally to the upper nasal passages. Serial blood samples were collected for 45 min, after which the animals were euthanized by transcardial perfusion-fixation. High resolution phosphor imaging of tissue sections and gamma counting of microdissected tissue were used to obtain the distribution and concentration profiles of [(125)I]-IFN-beta1b in central and peripheral tissues. Intranasal administration resulted in rapid, widespread targeting of nervous tissue. The olfactory bulbs and trigeminal nerve exhibited [(125)I]-IFN-beta1b levels significantly greater than in peripheral organs and at least one order of magnitude higher than any other nervous tissue area sampled. The basal ganglia exhibited highest [(125)I]-IFN-beta1b levels among CNS regions other than the olfactory bulbs. Preferential IFN-beta1b distribution to the primate basal ganglia is a new finding of possible clinical importance. Our study suggests both IFN-beta and IFN-alpha, which share the same receptor, may be bound with relatively high affinity in these structures, possibly offering new insight into a neurovegetative syndrome induced by IFN-alpha therapy and suspected to involve altered dopamine neurotransmission in the basal ganglia. Most importantly, our results suggest intranasally applied macromolecules may bypass the blood-brain barrier and rapidly enter the primate CNS along olfactory- and trigeminal-associated extracellular pathways, as shown previously in the rat. This is the first study to finely detail the central distribution of a labeled protein after intranasal administration in non-human primates.


Subject(s)
Interferon-beta/pharmacokinetics , Nervous System/drug effects , Olfactory Mucosa/drug effects , Administration, Intranasal , Animals , Autoradiography , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Mapping , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Extracellular Space/drug effects , Extracellular Space/metabolism , Immunologic Factors/administration & dosage , Immunologic Factors/metabolism , Immunologic Factors/pharmacokinetics , Interferon-beta/administration & dosage , Interferon-beta/metabolism , Iodine Radioisotopes , Macaca , Male , Nervous System/immunology , Nervous System/metabolism , Olfactory Bulb/drug effects , Olfactory Bulb/metabolism , Olfactory Mucosa/metabolism , Protein Binding/drug effects , Protein Binding/physiology , Radioimmunoassay , Receptors, Interferon/drug effects , Receptors, Interferon/metabolism , Trigeminal Nerve/drug effects , Trigeminal Nerve/metabolism
6.
Hepatogastroenterology ; 55(85): 1274-9, 2008.
Article in English | MEDLINE | ID: mdl-18795672

ABSTRACT

BACKGROUND/AIMS: The receptor of interferon-gamma (IFN-gammaR) consists of IFN-gammaR1 and R2. Resistance to the anti-proliferative effect of IFN-gamma is due to downregulation of IFN-gammaR2. The aim of this study was to investigate whether iron chelation could upregulate IFN-gammaR2 and enhance the anti-proliferative effect of IFN-gamma in colon cancer cell lines. METHODOLOGY: The colon cancer cell lines, SW480, COLO, and WiDr were treated with the iron chelating agent DFO, and the expression of IFN-gammaR1 and IFN-gammaR2 was evaluated by FACS. The anti-proliferative effect of IFN-gamma was investigated by MTT assay, and the proapoptotic effect was investigated by FACS with Annexin-V. RESULTS: FACS demonstrated that DFO increased the expression of IFN-gammaR2, whereas the effect on IFN-gammaR1 expression was less marked. MTT assay showed that cell growth was inhibited by DFO. Addition of DFO and IFN-gamma inhibited further, but inhibition was not observed with IFN-gamma alone. Apoptotic cells were increased by DFO, and further increased with DFO + IFN-gamma together. CONCLUSIONS: Expression of IFN-gammaR2 is restored by iron chelation, and the increased expression of IFN-gammaR2 enhances the anti-proliferative effect of IFN-gamma through induction of apoptosis in colon cancer cells.


Subject(s)
Carcinoma/metabolism , Colonic Neoplasms/metabolism , Deferoxamine/pharmacology , Receptors, Interferon/drug effects , Receptors, Interferon/metabolism , Siderophores/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma/pathology , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Humans , Interferon-gamma/pharmacology , Interferon gamma Receptor
7.
Acta Pol Pharm ; 65(3): 345-51, 2008.
Article in English | MEDLINE | ID: mdl-18646554

ABSTRACT

Interferon-alpha and ribavirin are currently the only drugs registered in the chronic hepatitis C therapy. Their actions are based on both direct antiviral activities, and their influence on genes expressions. In the presented study, using Jurkat cell line as an in vitro model for interferon-gamma synthesis, influence of interferon-alpha and ribavirin on the expressions of IFN-gamma and its receptor subunits (IFNgR1 and IFNgR2) were studied. Expressions of the studied genes were measured at the transcriptional level using Real-Time RT-PCR method. Results indicate that both drugs, IFN-alpha and ribavirin, induced changes in IFN-gamma and its receptor expressions. While IFN-alpha stimulated the expressions of the studied genes (IFN- gamma, IFNgR1, and IFNgR2), ribavirin showed the contradictory influence. The inhibitory effect of ribavirin dominated IFN-alpha action and was responsible for the decrease in the mRNA levels of IFN-gamma and the receptor of IFN-gamma. This phenomenon observed at in vitro model may be responsible for the IFN-gamma decrease during hepatitis C therapy with IFN-alpha and ribavirin which was suggested by some authors.


Subject(s)
Antiviral Agents/pharmacology , Interferon-alpha/pharmacology , Interferon-gamma/drug effects , Receptors, Interferon/drug effects , Ribavirin/pharmacology , Gene Expression Regulation/drug effects , Humans , Interferon-gamma/genetics , Jurkat Cells , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Interferon/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/drug effects , Interferon gamma Receptor
8.
Mol Cell Biol ; 15(8): 4208-14, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7623815

ABSTRACT

Two transmembrane polypeptides, IFNAR and IFN-alpha/Beta R, were previously identified as essential components of the type I interferon (IFN) receptor, but their interrelationship and role in ligand binding were not clear. To study these issues, we stably expressed and characterized the two polypeptides in host murine cells. In human cells, native IFN-alpha/beta R is a 102-kDa protein but upon reduction only a 51-kDa protein is detected. In host murine cells human IFN-alpha/beta R was expressed as a 51-kDa protein. Host cells expressing IFN-alpha/beta R bound IFN-alpha 2 with a high affinity (Kd of 3.6 nM), whereas cells expressing IFNAR exhibited no ligand binding. Upon coexpression of IFNAR and the 51-kDa IFN-alpha/beta R, the affinity for IFN-alpha 2 was increased 10-fold, approaching that of the native receptor. We show by cross-linking that both the cloned (51-kDa) and native (102-kDa) IFN-alpha/beta R bind IFN-alpha 2 to form an intermediate product, while IFNAR associates with this product to form a ternary complex. Hence, IFNAR and IFN-alpha/beta R are components of a common type I IFN receptor, cooperating in ligand binding. Ligand-induced association of IFNAR and IFN-alpha/beta R probably triggers transmembrane signaling.


Subject(s)
Interferon-alpha/pharmacology , Receptors, Interferon/drug effects , 3T3 Cells , Animals , Base Sequence , Cross-Linking Reagents , Humans , Interferon alpha-2 , Ligands , Membrane Proteins , Mice , Molecular Sequence Data , Precipitin Tests , Protein Binding , Receptor, Interferon alpha-beta , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism
9.
Mol Cell Biol ; 16(6): 3214-21, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8649432

ABSTRACT

Functionally active gamma interferon (IFN-gamma) receptors consist of an alpha subunit required for ligand binding and signal transduction and a beta subunit required primarily for signaling. Although the receptor alpha chain has been well characterized, little is known about the specific role of the receptor beta chain in IFN-gamma signaling. Expression of the wild-type human IFN-gamma receptor beta chain in murine L cells that stably express the human IFN-gamma receptor alpha chain (L.hgR) produced a murine cell line (L.hgR.myc beta) that responded to human IFN-gamma. Mutagenesis of the receptor beta-chain intracellular domain revealed that only two closely spaced, membrane-proximal sequences (P263PSIP267 and I270EEYL274) are required for function. Coprecipitation studies showed that these sequences are necessary for the specific and constitutive association of the receptor beta chain with the JAK-2 tyrosine kinase. These experiments also revealed that the IFN-gamma receptor alpha and beta chains are not preassociated on the surface of unstimulated cells but rather are induced to associate in an IFN-gamma-dependent fashion. A chimeric protein in which the intracellular domain of the beta chain was replaced by JAK-2 complemented human IFN-gamma signaling and biologic responsiveness in L.hgR. In contrast, a c-src-containing beta-chain chimera did not. These results indicate that the sole obligate role of the IFN-gamma receptor beta chain in signaling is to recruit JAK-2 into the ligand-assembled receptor complex.


Subject(s)
Antigens, CD/metabolism , Proto-Oncogene Proteins , Receptors, Interferon/metabolism , Amino Acid Sequence , Animals , Antigens, CD/drug effects , Antigens, CD/genetics , Binding Sites/genetics , Humans , Interferon-gamma/pharmacology , Janus Kinase 2 , L Cells , Ligands , Mice , Models, Biological , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Protein-Tyrosine Kinases/metabolism , Receptors, Interferon/drug effects , Receptors, Interferon/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/immunology , Interferon gamma Receptor
10.
Mol Cell Biol ; 20(13): 4724-35, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10848598

ABSTRACT

Hematopoietic progenitor cells from Fanconi anemia (FA) group C (FA-C) patients display hypersensitivity to the apoptotic effects of gamma interferon (IFN-gamma) and constitutively express a variety of IFN-dependent genes. Paradoxically, however, STAT1 activation is suppressed in IFN-stimulated FA cells, an abnormality corrected by transduction of normal FANCC cDNA. We therefore sought to define the specific role of FANCC protein in signal transduction through receptors that activate STAT1. Expression and phosphorylation of IFN-gamma receptor alpha chain (IFN-gammaRalpha) and JAK1 and JAK2 tyrosine kinases were equivalent in both normal and FA-C cells. However, in coimmunoprecipitation experiments STAT1 did not dock at the IFN-gammaR of FA-C cells, an abnormality corrected by transduction of the FANCC gene. In addition, glutathione S-transferase fusion genes encoding normal FANCC but not a mutant FANCC bearing an inactivating point mutation (L554P) bound to STAT1 in lysates of IFN-gamma-stimulated B cells and IFN-, granulocyte-macrophage colony-stimulating factor- and stem cell factor-stimulated MO7e cells. Kinetic studies revealed that the initial binding of FANCC was to nonphosphorylated STAT1 but that subsequently the complex moved to the receptor docking site, at which point STAT1 became phosphorylated. The STAT1 phosphorylation defect in FA-C cells was functionally significant in that IFN induction of IFN response factor 1 was suppressed and STAT1-DNA complexes were not detected in nuclear extracts of FA-C cells. We also determined that the IFN-gamma hypersensitivity of FA-C hematopoietic progenitor cells does not derive from STAT1 activation defects because granulocyte-macrophage CFU and erythroid burst-forming units from STAT1(-/-) mice were resistant to IFN-gamma. However, BFU-E responses to SCF and erythropoietin were suppressed in STAT(-/-) mice. Consequently, because the FANCC protein is involved in the activation of STAT1 through receptors for at least three hematopoietic growth and survival factor molecules, we reason that FA-C hematopoietic cells are excessively apoptotic because of an imbalance between survival cues (owing to a failure of STAT1 activation in FA-C cells) and apoptotic and mitogenic inhibitory cues (constitutively activated in FA-C cells in a STAT1-independent fashion).


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins/metabolism , Growth Substances/metabolism , Interferon-gamma/metabolism , Nuclear Proteins , Proteins/metabolism , Proto-Oncogene Proteins , Trans-Activators/metabolism , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Line/drug effects , DNA-Binding Proteins/genetics , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group C Protein , Fanconi Anemia Complementation Group Proteins , Growth Substances/pharmacology , Humans , Interferon Regulatory Factor-1 , Interferon-gamma/pharmacology , Janus Kinase 1 , Janus Kinase 2 , Mice , Mice, Knockout , Mutation , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Precipitin Tests , Protein-Tyrosine Kinases/metabolism , Proteins/genetics , Receptors, Interferon/drug effects , Receptors, Interferon/metabolism , STAT1 Transcription Factor , Trans-Activators/genetics , Interferon gamma Receptor
11.
Mol Cell Biol ; 20(19): 7121-31, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10982828

ABSTRACT

Stat4 is activated by the cytokines interleukin 12 and alpha interferon (IFN-alpha) and plays a significant role in directing development of naïve CD4(+) T cells to the Th1 phenotype. Signal transducers and activators of transcription (STAT) proteins undergo phosphorylation on a conserved tyrosine residue, resulting in homo- and heterodimerization, nuclear translocation, and DNA binding. Stat4 can bind to single IFN-gamma-activated sites (GASs) as a dimer or bind two tandem GASs as a pair of STAT dimers, or tetramer, stabilized through N-terminal domain (N domain) interactions between dimers. We uncovered an unexpected effect of the Stat4 N domain in controlling the proximal activation of Stat4 by tyrosine phosphorylation at activated receptor complexes. Mutation of the N domain at tryptophan residue W37, predicted to interrupt N domain dimer formation, unexpectedly prevented IFN-alpha-induced tyrosine phosphorylation of the Stat4 monomer, blocking dimer formation and nuclear translocation. Furthermore, N domains appear to exert private STAT functions, since interchanging the N domains between Stat1 and Stat4 prevented receptor-mediated tyrosine phosphorylation in one case and interrupted STAT-specific gene activation in another. Finally, replacement of the N domain of Stat1 with that of Stat4 abrogated the normal Stat2 dependence of Stat1 phosphorylation, again suggesting the domains are not equivalent. Thus, in addition to its role in STAT tetramerization, the conserved STAT N domain appears to participate in very proximal steps of receptor-mediated ligand-induced tyrosine phosphorylation.


Subject(s)
DNA-Binding Proteins/physiology , Phosphotyrosine/biosynthesis , Protein Processing, Post-Translational , Trans-Activators/physiology , Tyrosine/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Cells, Cultured , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dimerization , Humans , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Ligands , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/metabolism , Receptor, Interferon alpha-beta , Receptors, Interferon/drug effects , Receptors, Interferon/metabolism , Receptors, Interleukin-2/drug effects , Receptors, Interleukin-2/physiology , Recombinant Fusion Proteins/physiology , Recombinant Proteins/pharmacology , STAT1 Transcription Factor , STAT4 Transcription Factor , Structure-Activity Relationship , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription, Genetic
12.
Am J Surg ; 191(3): 358-63, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16490547

ABSTRACT

BACKGROUND: Interferons (IFNs) have antiproliferative effects on tumor cells. The apoptotic effects and sensitization to chemotherapy conferred by IFN therapy, however, are not clearly understood. The aims of the present study were to explore the apoptotic effects of IFNs in human pancreatic cancer cell lines and to attempt to define their ability to synergistically enhance sensitivity to 5-fluorouracil (5-FU) and gemcitabine, a mechanism that depends on the expression of IFN receptors. METHODS: Human pancreatic cancer cells were cultured alone or in combination with the chemotherapeutic agents 5-FU and gemcitabine. Differential dosages of IFN-alpha, -beta, and -gamma were also added to the cell lines concomitantly during a period of 24 to 96 hours. The cell line viability and effects of treatment were examined using the methylthiazol tetrazolium assay and single-stranded DNA apoptosis assay. The expression of IFN receptors was determined using immunohistochemistry. Caspase-8 inhibitor was used to block the caspase cascade. RESULTS: The antiproliferative and apoptotic effects of IFNs were most profoundly demonstrated on those cells that expressed the respective IFN receptor. The apoptotic effects provided by the interferons, however, were blocked by caspase-8 inhibition. The addition of IFNs significantly enhanced the cytotoxic effects of 5-FU and gemcitabine in those cell lines that expressed the corresponding IFN-alpha, -beta, or -gamma receptors. CONCLUSIONS: This study on pancreatic cancer cell lines has demonstrated that IFNs mediate apoptosis through IFN receptors and the caspase cascade. Enhanced cytotoxicity occurred when IFNs were combined with 5-FU and gemcitabine.


Subject(s)
Antineoplastic Agents/pharmacology , Caspases/drug effects , Interferons/pharmacology , Pancreatic Neoplasms/drug therapy , Receptors, Interferon/drug effects , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Caspase Inhibitors , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Synergism , Fluorouracil/pharmacology , Humans , Interferons/administration & dosage , Gemcitabine
13.
J Natl Cancer Inst ; 86(22): 1694-701, 1994 Nov 16.
Article in English | MEDLINE | ID: mdl-7966397

ABSTRACT

BACKGROUND: Interferon gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF) synergize in inducing human neuroblastoma cells to differentiate terminally in vitro into mature nonproliferating neurons. The mechanisms by which this synergistic activity takes place are still obscure. PURPOSE: To understand the basis of IFN-gamma-TNF synergism, we investigated the constitutive equipment of receptors to IFN-gamma and TNF in two human neuroblastoma cell lines (i.e., LAN-5 and GI-LI-N) and their quantitative and functional variations following treatment with IFN-gamma or TNF. METHODS: IFN-gamma receptors and TNF receptors were assessed and functionally characterized by radioreceptor-binding assay before and after treatment of the cells with IFN-gamma or TNF. The TNF receptor subtypes were identified by the reverse transcriptase-polymerase chain reaction, chemical cross-linking of receptors to iodinated TNF, and inhibition of TNF binding by type-specific anti-TNF receptor monoclonal antibodies. The effects of cytokines on cell differentiation were assessed by thymidine incorporation inhibition and morphologic maturation. RESULTS: No quantitative or functional modification of IFN-gamma receptors was observed in TNF-treated cells. However, after treatment with IFN-gamma, TNF receptor numbers were enhanced to a different extent in both cell lines. The two neuroblastoma cell lines expressed, both constitutively and after IFN-gamma induction, only one species of TNF receptor, i.e., the p80 form in LAN-5 and the p60 form in GI-LI-N. Sequential treatment with IFN-gamma followed by TNF, but not in the opposite order, could reproduce the early effects of differentiation in neuroblastoma cells, supporting a role for TNF receptor up-regulation as a basis for the cooperation between the two cytokines. CONCLUSION: The results strongly suggest that receptor regulation can be at least one mechanism by which IFN-gamma and TNF exert their synergistic effects. Moreover, it appears that the two TNF receptor types are redundant in signaling neuroblastoma cell differentiation. IMPLICATIONS: Our findings can provide a guideline for a rational design of experimental differentiation-based therapeutic protocols in patients with neuroblastoma.


Subject(s)
Interferon-gamma/pharmacology , Neuroblastoma/metabolism , Receptors, Interferon/drug effects , Receptors, Tumor Necrosis Factor/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Base Sequence , Drug Synergism , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
14.
Cancer Res ; 53(3): 636-40, 1993 Feb 01.
Article in English | MEDLINE | ID: mdl-8425199

ABSTRACT

Recombinant human tumor necrosis factor and recombinant human gamma interferon (IFN-gamma) exert synergistic growth inhibitory effects in WiDR human colorectal carcinoma cells. In this cell line, tumor necrosis factor increases IFN-gamma binding. Interleukin 1 (IL-1) is a cytokine that mimics many of the biological actions of TNF. Therefore, in the present study, we investigated the effects of recombinant human IL-1 on cell growth and IFN-gamma receptor expression in WiDR cells. IL-1 slightly inhibited the growth of WiDR cells, and exerted additive growth inhibitory effects in the presence of IFN-gamma. IL-1 caused a time- and dose-dependent increase in 125I-labeled IFN-gamma binding that was maximal at 6 h, persisted for at least 24 h, and was blocked by both actinomycin D and cycloheximide. The increase in binding was associated with an increase in cell surface IFN-gamma receptor protein expression as determined by Scatchard analysis of equilibrium binding data and by immunofluorescent staining with an anti-human IFN-gamma receptor monoclonal antibody. IL-1 also produced a time- and dose-dependent increase in IFN-gamma receptor mRNA levels that was maximal at 3 h and persisted for at least 24 h. Actinomycin D, but not cycloheximide, completely blocked the IL-1-mediated increase in IFN-gamma receptor mRNA levels. However, IL-1 did not alter IFN-gamma receptor mRNA half-life. These data indicate that IL-1 and IFN-gamma exert additive growth inhibitory effects on colon cancer cell growth, and suggest that IL-1 increases IFN-gamma receptor expression in these cells by enhancing IFN-gamma mRNA levels.


Subject(s)
Colorectal Neoplasms/drug therapy , Interleukin-1/pharmacology , Receptors, Interferon/physiology , Cell Division/drug effects , Colorectal Neoplasms/pathology , Colorectal Neoplasms/ultrastructure , Humans , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Iodine Radioisotopes , Kinetics , Neoplasm Proteins/biosynthesis , RNA, Messenger/metabolism , RNA, Neoplasm/biosynthesis , Receptors, Interferon/drug effects , Receptors, Interferon/genetics , Recombinant Proteins , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology , Interferon gamma Receptor
15.
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
16.
Biochim Biophys Acta ; 1294(1): 55-62, 1996 May 02.
Article in English | MEDLINE | ID: mdl-8639714

ABSTRACT

A gene encoding a 195 amino-acid (a.a.) polypeptide with a putative 23 a.a. signal sequence that had about 60% a.a. sequence identity to ovine interferon-omega (OvIFN-omega) and 55% or less identity to BoIFN-tau, OvIFN-tau and all known IFN-alpha and -beta has been identified from an ovine genomic DNA library. Surprisingly, it shared almost complete identity to genes for rabbit IFN-omega within its coding sequence and proximal promoter region, although the two were different in their 3'-ends. This IFN (tentatively termed ovine IFN-omega variant, OvIFN-omegav), purified in recombinant form from E. coli, had normal antiviral activity when tested on sheep fetal tongue and brain cells and rabbit kidney cells, but very low activity towards bovine, goat and human cells. It competed with 125I-labeled BoIFN-tau for binding to IFN receptors on ovine cells. Expression of OvIFN-omegav was not detected by reverse transcription-PCR either in ovine peripheral blood leukocytes infected with Sendai virus, or in any other tissues examined. OvIFN-omegav may represent a previously unrecognized, non-virally inducible type I subtype distinct from IFN-alpha, -beta, -omega and -tau. The presence of a conserved gene in rabbit and sheep could reflect a recent interspecies transfer.


Subject(s)
Interferon Type I/genetics , Interferon Type I/pharmacology , Amino Acid Sequence , Animals , Antiviral Agents/pharmacology , Base Sequence , Binding, Competitive , Blotting, Western , Cell Division/drug effects , Cell Line , Cloning, Molecular , DNA Primers/chemistry , Interferon Type I/chemistry , Interferon Type I/metabolism , Interferon-alpha/chemistry , Molecular Sequence Data , Pregnancy Proteins/chemistry , Receptors, Interferon/drug effects , Receptors, Interferon/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Sequence Alignment , Sheep , Species Specificity
17.
Diabetes ; 46(5): 770-8, 1997 May.
Article in English | MEDLINE | ID: mdl-9133543

ABSTRACT

The mouse pancreatic beta TC3 and beta TC6-F7 cell lines were used to characterize the effects of interferon-gamma (IFN-y) on beta-cell phenotype and function. Initially, intracellular and secreted insulin were compared in glucose-stimulated cells over time. A significant reduction in insulin content and secretion was observed on a per-cell basis in glucose-stimulated beta TC3 and beta TC6-F7 cells after 12 h of exposure to IFN-gamma. The steadystate level of pre-proinsulin mRNA expression was not affected by IFN-gamma. Thus, we postulate that IFN-gamma's inhibitory actions occur after transcription of pre-proinsulin genes. Time-course analysis of IFN-gamma-regulated mRNA expression of the two intra-MHC-encoded subunits of the proteasome (low-molecular-mass polypeptide [Lmp]-2 and Lmp-7) revealed a correlation between their induction and the inhibitory effects of IFN-gamma on glucose-stimulated insulin production. Increased expression of Lmp-2 and Lmp-7 mRNA was accompanied by a corresponding induction of LMP2 and LMP7 protein expression. Subsequently, major histocompatibility complex (MHC) class I cell-surface expression was significantly increased in IFN-gamma-treated beta TC3 and beta TC6-F7 cells. Exposure of IFN-gamma-treated beta-cells to a peptide aldehyde inhibitor of the proteasome (MG132) significantly attenuated MHC class I cell-surface expression but did not prevent the negative effects of IFN-gamma on glucose responsiveness. Enhanced expression of the MHC class I antigen processing and presentation pathway and diminished insulin production appear to be distinct pathological alterations in beta-cells exposed to the insulitic cytokine IFN-gamma.


Subject(s)
Cysteine Endopeptidases , Glucose/physiology , Histocompatibility Antigens Class I/immunology , Interferon-gamma/pharmacology , Islets of Langerhans/metabolism , Multienzyme Complexes , Animals , Antigen Presentation/drug effects , Blotting, Northern , Blotting, Western , Cysteine Proteinase Inhibitors/pharmacology , Insulin/analysis , Insulin/metabolism , Islets of Langerhans/chemistry , Leupeptins/pharmacology , Major Histocompatibility Complex/genetics , Mice , Proinsulin/genetics , Proteasome Endopeptidase Complex , Protein Precursors/genetics , Proteins/genetics , RNA/analysis , Receptors, Interferon/drug effects , Time Factors , Tumor Cells, Cultured/drug effects , Interferon gamma Receptor
18.
Pharmacol Ther ; 66(3): 507-34, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7494857

ABSTRACT

The alpha-interferons (IFN-alpha) belong to a family of polypeptides comprising several subtypes. Using recombinant DNA technology, it has been possible to create IFN hybrids that provide novel combinations of the amino acid residues from the parental protein sequences. They have been used to study structure-activity relationships of IFN-alpha and interactions with the IFN-alpha receptor, and to create analogs of natural IFNs with novel properties for potential therapeutic application. The biological data obtained with these hybrids are now evaluated in terms of the published structural and homology models of IFN-beta and -alpha.


Subject(s)
Interferon-alpha/pharmacology , Receptors, Interferon/drug effects , Animals , Cattle , Computer Simulation , DNA, Recombinant/chemistry , DNA, Recombinant/genetics , Gene Expression Regulation/genetics , Humans , Interferon-alpha/chemistry , Interferon-alpha/classification , Interferon-alpha/genetics , Interferon-alpha/metabolism , Interferon-alpha/therapeutic use , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Mice , Models, Structural , Mutation , Receptors, Interferon/metabolism , Structure-Activity Relationship , Terminology as Topic
19.
J Leukoc Biol ; 68(1): 119-24, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10914498

ABSTRACT

DBA/1 mice deficient in expressing the interferon-gamma (IFN-gamma) membrane receptor (IFN-gammaR KO mice) are more susceptible to collagen-induced arthritis (CIA) than wild-type mice, indicating that endogenous IFN-gamma plays a protective role in the pathogenesis of CIA. In IFN-gammaR KO mice, nitric oxide (NO) production during CIA is impaired. Because NO is known to exert immunosuppressive and anti-inflammatory effects in certain model systems, the protective effect of IFN-gamma might be mediated by NO. Here, we tested in wild-type mice whether inhibition of NO production by metabolic inhibitors, aminoguanidine (AG) and L-N-(1-iminoethyl)lysine (L-NIL), could mimic the ablation of the IFN-gamma receptor. A high-dose regimen of AG supplied in the drinking water inhibited NO production, disease development, and anticollagen antibody production but was also associated with transient body weight loss. At a dose and time regimen that still inhibited NO production but did not cause body weight loss, AG failed to affect disease scores. Treatment with L-NIL, which more specifically than AG affects inducible NO production, caused a slight increase in anticollagen antibody production although not significantly affecting disease occurrence. These data indicate that the diminished capacity of the IFN-gammaR KO mice to produce NO following immunization with collagen is unlikely to account for their higher susceptibility to CIA.


Subject(s)
Arthritis, Rheumatoid/physiopathology , Autoimmune Diseases/physiopathology , Collagen/toxicity , Enzyme Inhibitors/therapeutic use , Guanidines/therapeutic use , Interferon-gamma/physiology , Lysine/analogs & derivatives , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/physiology , Administration, Oral , Animals , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/prevention & control , Autoantibodies/biosynthesis , Autoantibodies/immunology , Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Collagen/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Female , Genetic Predisposition to Disease , Guanidines/administration & dosage , Guanidines/pharmacology , Lysine/administration & dosage , Lysine/pharmacology , Lysine/therapeutic use , Male , Mice , Mice, Inbred DBA , Mice, Knockout , Nitrates/blood , Nitric Oxide Synthase Type II , Nitrites/blood , Receptors, Interferon/deficiency , Receptors, Interferon/drug effects , Receptors, Interferon/genetics , Weight Loss/drug effects , Interferon gamma Receptor
20.
J Leukoc Biol ; 76(6): 1171-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15339939

ABSTRACT

Natural killer (NK) cells are critical in host innate defense against certain viruses. The role of NK cells in controlling hepatitis C virus (HCV) remains obscure. We examined whether NK cells are capable of inhibiting HCV expression in human hepatic cells. When NK cells are cultured with the HCV replicon-containing hepatic cells, they have no direct cytolytic effect but release soluble factor(s) suppressing HCV RNA expression. Media conditioned by NK cell lines (NK-92 and YTS) or primary NK cells isolated from healthy donors contain interferon gamma (IFN-gamma) and potently inhibit HCV RNA expression. Ligation of CD81 on NK cells inhibits IFN-gamma production and results in decreased anti-HCV activity. In addition, the antibodies to IFN-gamma or IFN-gamma receptors abolish the anti-HCV activity of NK cell-conditioned media. The role of IFN-gamma in NK cell-mediated, anti-HCV activity is supported by the observation that NK cell-conditioned media enhanced expression of signal transducer and activator of transcription-1, a nuclear factor that is essential in IFN-gamma-mediated antiviral pathways. NK cell-conditioned media have the ability to stimulate intracellular IFN-alpha expression in the hepatic cells, suggesting a mechanism responsible for NK cell-mediated, anti-HCV activity. Thus, NK cells hold the potential to play a vital role in controlling HCV replication in hepatic cells using an IFN-gamma-dependent mechanism.


Subject(s)
Antiviral Agents/immunology , Hepacivirus/immunology , Immunity, Innate/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Virus Replication/immunology , Antigens, CD/drug effects , Antigens, CD/immunology , Antiviral Agents/metabolism , Biological Assay , Cell Line , Coculture Techniques , Culture Media, Conditioned/pharmacology , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Down-Regulation/immunology , Hepacivirus/drug effects , Hepatocytes/immunology , Hepatocytes/virology , Humans , Interferon-alpha/immunology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/virology , RNA, Viral/biosynthesis , Receptors, Interferon/drug effects , Receptors, Interferon/immunology , Repressor Proteins/immunology , Repressor Proteins/metabolism , STAT1 Transcription Factor , Tetraspanin 28 , Trans-Activators/immunology , Trans-Activators/metabolism , Virus Replication/drug effects , Interferon gamma Receptor
SELECTION OF CITATIONS
SEARCH DETAIL