Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cancer Immunol Immunother ; 68(3): 395-405, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30552459

ABSTRACT

Optimal ex vivo expansion protocols for adoptive cell therapy (ACT) must yield T cells able to effectively home to tumors and survive the inhospitable conditions of the tumor microenvironment (TME), while simultaneously exerting persistent anti-tumor effector functions. Our previous work has shown that ex vivo activation in the presence of IL-12 can induce optimal expansion of murine CD8+ T cells, thus resulting in significant tumor regression after ACT mostly via sustained secretion of IFN-γ. In this report, we further elucidate the mechanism of this potency, showing that IL-12 additionally counteracts the negative regulatory effects of autocrine IFN-γ. IL-12 not only downregulates PD-1 expression by T cells, thus minimizing the effects of IFN-γ-induced PD-L1 upregulation by tumor stromal cells, but also inhibits IFNγR2 expression, thereby protecting T cells from IFN-γ-induced cell death. Thus, the enhanced anti-tumor activity of CD8+ T cells expanded ex vivo in the presence of IL-12 is due not only to the ability of IL-12-stimulated cells to secrete sustained levels of IFN-γ, but also to the additional capacity of IL-12 to counter the negative regulatory effects of autocrine IFN-γ.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Cytotoxicity, Immunologic/drug effects , Interferon-gamma/physiology , Interleukin-12/pharmacology , Lymphocytes, Tumor-Infiltrating/drug effects , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Humans , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Receptors, Interferon/analysis , Receptors, Interferon/physiology , Interferon gamma Receptor
2.
J Immunol ; 188(3): 1011-8, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22219326

ABSTRACT

IFN-ß inhibits the expansion of Th17 cells in active multiple sclerosis (AMS), and this might contribute to improve the clinical symptoms. The effectiveness of this inhibition, however, requires intact IFN-γ signaling in T cells. In this study, we report that both mRNA and cell surface expression of the signaling chain of the IFN-γ receptor (IFN-γR2) and its cognate tyrosine kinase JAK2 are enhanced in peripheral blood Th17 cells and clones from patients with AMS compared with those with inactive multiple sclerosis (IMS) or healthy subjects (HS). IFN-γ decreased the frequency of Th17 peripheral cells and proliferation of Th17 clones from AMS patients. Stimulation of PBMCs from HS in Th17-polarizing conditions resulted in the enhancement of JAK2 expression and accumulation of cell surface IFN-γR2. The role of JAK2 in the modulation of IFN-γR2 was demonstrated as its transduction prevented rapid internalization and degradation of IFN-γR2 in JAK2-deficient γ2A cells. In conclusion, these data identify JAK2 as a critical factor that stabilizes IFN-γR2 surface expression in Th17 cells from AMS patients, making them sensitive to IFN-γ. These data may have clinical implications for a better use of IFNs in multiple sclerosis and possibly other inflammatory diseases.


Subject(s)
Janus Kinase 2/metabolism , Multiple Sclerosis/immunology , Receptors, Interferon/metabolism , Th17 Cells/metabolism , Case-Control Studies , Cell Proliferation , Humans , Interferons , Multiple Sclerosis/pathology , RNA, Messenger/analysis , Receptors, Interferon/analysis , Th17 Cells/immunology , Interferon gamma Receptor
3.
Cytokine ; 64(1): 272-85, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23796694

ABSTRACT

The observed Fluorescence Resonance Energy Transfer (FRET) between fluorescently labeled proteins varies in cells. To understand how this variation affects our interpretation of how proteins interact in cells, we developed a protocol that mathematically separates donor-independent and donor-dependent excitations of acceptor, determines the electromagnetic interaction of donors and acceptors, and quantifies the efficiency of the interaction of donors and acceptors. By analyzing large populations of cells, we found that misbalanced or insufficient expression of acceptor or donor as well as their inefficient or reversible interaction influenced FRET efficiency in vivo. Use of red-shifted donors and acceptors gave spectra with less endogenous fluorescence but produced lower FRET efficiency, possibly caused by reduced quenching of red-shifted fluorophores in cells. Additionally, cryptic interactions between jellyfish FPs artefactually increased the apparent FRET efficiency. Our protocol can distinguish specific and nonspecific protein interactions even within highly constrained environments as plasma membranes. Overall, accurate FRET estimations in cells or within complex environments can be obtained by a combination of proper data analysis, study of sufficient numbers of cells, and use of properly empirically developed fluorescent proteins.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Janus Kinases/analysis , Receptors, Interferon/analysis , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Fluorescent Dyes/metabolism , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Interferon-gamma/metabolism , Luminescent Proteins/metabolism , Microscopy, Fluorescence/methods , Multiprotein Complexes , Receptors, Interferon/metabolism , Staining and Labeling , Interferon gamma Receptor , Red Fluorescent Protein
4.
J Exp Med ; 181(5): 1893-8, 1995 May 01.
Article in English | MEDLINE | ID: mdl-7722464

ABSTRACT

Interleukin (IL)-12 synergizes with other cytokines to stimulate the proliferation and differentiation of early hematopoietic progenitors in vitro. However, in vivo administration of IL-12 decreases peripheral blood counts and bone marrow hematopoiesis. Here, we used interferon (IFN) gamma receptor-deficient (IFN gamma R-/-) mice to investigate whether the in vivo inhibition of hematopoiesis by IL-12 is indirectly mediated by IL-12-induced IFN-gamma. IL-12 administered for 4 d (1 microgram/mouse per day) resulted in lower peripheral blood counts and a 2-fold decrease in bone marrow cellularity in wild-type mice, but not in IFN gamma R-/- mice. Bone marrow hematopoietic progenitors were decreased after IL-12 treatment in wild-type mice, but rather increased in IFN gamma R-/- mice. Splenic cellularity was 2.3-fold higher after IL-12 administration in wild-type mice, largely due to natural killer (NK) cell and macrophage infiltration together with some extramedullary hematopoiesis. In IFN gamma R-/- mice, spleen cellularity was less increased, there were fewer infiltrating NK cells, but a strong extramedullary hematopoiesis. Thus, alterations mediated by IL-12-induced IFN-gamma include reduction in bone marrow cellularity and hematopoietic progenitors, as well as pronounced splenomegaly, largely caused by NK cell infiltration. In the absence of IFN-gamma signaling, IL-12 promotes hematopoiesis, consistent with its in vitro activities.


Subject(s)
Hematopoiesis/drug effects , Interferon-gamma/physiology , Interleukin-12/antagonists & inhibitors , Animals , Bone Marrow/drug effects , Mice , Receptors, Interferon/analysis , Interferon gamma Receptor
5.
J Exp Med ; 185(3): 517-30, 1997 Feb 03.
Article in English | MEDLINE | ID: mdl-9053452

ABSTRACT

The hematologic consequences of infection with the noncytopathic lymphocytic choriomeningitis virus (LCMV) were studied in wild-type mice with inherent variations in their interferon (IFN)-alpha/beta responder ability and in mutant mice lacking alpha/beta (IFN-alpha/beta R0/0) or gamma IFN (IFN-gamma R0/0) receptors. During the first week of infection, wild type mice demonstrated a transient pancytopenia. Within a given genetic background, the extent of the blood cell abnormalities did not correlate with the virulence of the LCMV isolate but variations were detected between different mouse strains: they were found to depend on their IFN-alpha/beta responder phenotype. Whereas IFN-gamma R0/0 mice were comparable to wild-type mice, IFN-alpha/beta R0/0 mice exhibited unchanged peripheral blood values during acute LCMV infection. In parallel, the bone marrow (BM) cellularity, the pluripotential and committed progenitor compartments were up to 30-fold reduced in wild type and IFN-gamma R0/0, but remained unchanged in IFN-alpha/beta R0/0 mice. Viral titers in BM 3 d after LCMV infection were similar in these mice, but antigen localization was different. Viral antigen was predominantly confined to stromal BM in normal mice and IFN-gamma R0/0 knockouts, whereas, in IFN-alpha/beta R0/0 mice, LCMV was detected in > 90% of megakaryocytes and 10-15% of myeloid precursors, but not in erythroblasts Although IFN-alpha/beta efficiently prevented viral replication in potentially susceptible hematopoietic cells, even in overwhelming LCMV infection, unlimited virus multiplication in platelet and myeloid precursors in IFN-alpha/beta R0/0 mice did not interfere with the number of circulating blood cells. Natural killer (NK) cell expansion and activity in the BM was comparable on day 3 after infection in mutant and control mice. Adaptive immune responses did not play a major role because comparable kinetics of LCMV-induced pancytopenia and transient depletion of the pluripotential and committed progenitor compartments were observed in CD8(0/0) and CD4(0/0) mice, in mice depleted of NK cells, in lpr mice, and in perforin-deficient (P0/0) mice lacking lytic NK cells. Thus, the reversible depression of hematopoiesis during early LCMV infection was not mediated by LCMV-WE-specific cytotoxic T lymphocyte, cytolysis, or secreted IFN-gamma from virally induced NK cells but was a direct effect of IFN-alpha/beta.


Subject(s)
Bone Marrow/physiology , Interferon-alpha/physiology , Interferon-beta/physiology , Lymphocytic Choriomeningitis/blood , Acute Disease , Animals , Blood Cell Count , Cytotoxicity, Immunologic , Hematopoiesis , Hematopoietic Stem Cells , Killer Cells, Natural/immunology , Lymphocyte Activation , Mice , Mice, Inbred Strains , Receptors, Interferon/analysis , Virus Replication
6.
J Exp Med ; 181(6): 2069-76, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7759999

ABSTRACT

The Daniels strain of Theiler's virus causes a persistent infection of the white matter of spinal cord of susceptible mice, with chronic inflammation and primary demyelination. Inbred 129Sv mice are resistant to this infection; they present with mild encephalomyelitis and clear the infection within a matter of days. A very different outcome was observed with inbred 129Sv mice whose receptors for interferon alpha/beta or interferon gamma had been inactivated by homologous recombination. The former presented severe encephalomyelitis with acute infection of neurons, particularly in brain and hippocampus, and extensive infection with necrosis of the choroid plexus. Most animals died of this acute disease. The latter, presented the same early encephalomyelitis as the control 129Sv mice. However, they remained persistently infected and developed a very severe late infection of the white matter with extensive primary demyelination. This late disease looked like an exacerbated form of the chronic demyelinating disease observed in susceptible inbred mice such as the SJL/J or FVB strains. Our results show that the two interferon systems play nonredundant roles in the resistance of the 129Sv mouse to the infection by Theiler's virus. They also lend support to the notion that the Ifg gene is involved in the resistance/susceptibility of inbred strains of mice to persistent infection by this picornavirus.


Subject(s)
Brain/pathology , Poliomyelitis/pathology , Poliomyelitis/physiopathology , Receptors, Interferon/genetics , Spinal Cord/pathology , Theilovirus , Animals , Brain/immunology , Death , Immunohistochemistry , Membrane Proteins , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Poliomyelitis/immunology , Receptor, Interferon alpha-beta , Receptors, Interferon/analysis , Receptors, Interferon/biosynthesis , Recombination, Genetic , Spinal Cord/immunology , Time Factors , Interferon gamma Receptor
7.
J Exp Med ; 186(12): 2023-31, 1997 Dec 15.
Article in English | MEDLINE | ID: mdl-9396771

ABSTRACT

We explored expression and possible function of interferon-gamma (IFN-gamma) in cultured fetal (E15) rat dorsal root ganglion neurons combining whole cell patch-clamp electrophysiology with single cell reverse transcriptase polymerase chain reaction and confocal laser immunocytochemistry. Morphologically, we located IFN-gamma protein in the cytoplasm of the neurons in culture as well as in situ during peri- and postnatal development. Transcripts for classic IFN-gamma and for its receptor were determined in probes of cytoplasm sampled from individual cultured neurons, which had been identified by patch clamp electrophysiology. In addition, the cultured neurons expressed both chains of the IFN-gamma receptor. Locally produced IFN-gamma acts back on its cellular source. Phosphorylation and nuclear translocation of the IFN-inducible transcriptional factor STAT1 as well as IFN-gamma-dependent expression of major histocompatibility complex class I molecules on the neuronal membrane were noted in untreated cultures. However, both processes were substantially blocked in the presence of antibodies neutralizing IFN-gamma. Our findings indicate a role of IFN-gamma in autocrine regulation of sensory neurons.


Subject(s)
Gene Expression , Interferon-gamma/genetics , Neurons, Afferent/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Gene Expression Regulation, Developmental , Interferon-gamma/analysis , Microscopy, Phase-Contrast , Neurons, Afferent/chemistry , Polymerase Chain Reaction , Rats , Receptors, Interferon/analysis , Receptors, Interferon/metabolism , STAT1 Transcription Factor , Signal Transduction , Trans-Activators/metabolism , Transcription, Genetic , Interferon gamma Receptor
8.
J Immunoassay Immunochem ; 30(4): 378-85, 2009.
Article in English | MEDLINE | ID: mdl-19739012

ABSTRACT

We planned to investigate the urinary soluble cytokine receptor profile in patients with vesico-ureteric reflux (VUR). The urine levels of soluble interferon-gamma receptor R1 (sIFN-gammaR) and soluble interleukin-4 receptor alpha (sIL-4R) were measured using an ELISA technique. The urine levels of sIFN-gammaR in the patients with VUR were significantly higher than those in the healthy controls (p < 0.001). On the other hand, although the urine sIL-4R levels in the patients with VUR were also higher than those in the controls, there were no significant differences between them. The urinary soluble receptor levels did not correlate with the clinical severity of VUR. These results suggest that there may be an immunological basis to VUR complicatedly.


Subject(s)
Receptors, Interferon/analysis , Receptors, Interleukin-4/analysis , Vesico-Ureteral Reflux/urine , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infant , Male , Vesico-Ureteral Reflux/diagnosis , Vesico-Ureteral Reflux/immunology , Interferon gamma Receptor
9.
Pediatrics ; 144(3)2019 09.
Article in English | MEDLINE | ID: mdl-31455612

ABSTRACT

BACKGROUND: Diagnosis of tuberculosis should be improved in children infected with HIV to reduce mortality. We developed prediction scores to guide antituberculosis treatment decision in HIV-infected children with suspected tuberculosis. METHODS: HIV-infected children with suspected tuberculosis enrolled in Burkina Faso, Cambodia, Cameroon, and Vietnam (ANRS 12229 PAANTHER 01 Study), underwent clinical assessment, chest radiography, Quantiferon Gold In-Tube (QFT), abdominal ultrasonography, and sample collection for microbiology, including Xpert MTB/RIF (Xpert). We developed 4 tuberculosis diagnostic models using logistic regression: (1) all predictors included, (2) QFT excluded, (3) ultrasonography excluded, and (4) QFT and ultrasonography excluded. We internally validated the models using resampling. We built a score on the basis of the model with the best area under the receiver operating characteristic curve and parsimony. RESULTS: A total of 438 children were enrolled in the study; 251 (57.3%) had tuberculosis, including 55 (12.6%) with culture- or Xpert-confirmed tuberculosis. The final 4 models included Xpert, fever lasting >2 weeks, unremitting cough, hemoptysis and weight loss in the past 4 weeks, contact with a patient with smear-positive tuberculosis, tachycardia, miliary tuberculosis, alveolar opacities, and lymph nodes on the chest radiograph, together with abdominal lymph nodes on the ultrasound and QFT results. The areas under the receiver operating characteristic curves were 0.866, 0.861, 0.850, and 0.846, for models 1, 2, 3, and 4, respectively. The score developed on model 2 had a sensitivity of 88.6% and a specificity of 61.2% for a tuberculosis diagnosis. CONCLUSIONS: Our score had a good diagnostic performance. Used in an algorithm, it should enable prompt treatment decision in children with suspected tuberculosis and a high mortality risk, thus contributing to significant public health benefits.


Subject(s)
AIDS-Related Opportunistic Infections/diagnosis , Clinical Decision Rules , HIV Infections/complications , Tuberculosis/complications , Tuberculosis/diagnosis , AIDS-Related Opportunistic Infections/drug therapy , Abdomen/diagnostic imaging , Antitubercular Agents/therapeutic use , Bacteriological Techniques , Child , Child, Preschool , Female , Humans , Lung/diagnostic imaging , Male , Microscopy , Radiography , Receptors, Interferon/analysis , Reproducibility of Results , Sensitivity and Specificity , Tuberculosis/drug therapy , Ultrasonography , Interferon gamma Receptor
10.
Int J Parasitol ; 38(2): 249-58, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17822706

ABSTRACT

IFN-gamma production is a hallmark of acute infection with the protozoan parasite Toxoplasma gondii. The tryptophan-catabolising enzyme indoleamine 2,3-dioxygenase (IDO), as well as inducible nitric oxide synthase (NOS2) are induced by IFN-gamma and can play extremely diverse roles in immune regulation, defence against pathogens and physiological homeostasis. We investigated the regulation of these two central enzymes in the placenta during acute infection of pregnant female mice. Using IFN-gamma receptor knockout (IFNgammaR-/-) mice, we showed that IDO is not constitutively expressed in term placentas. In contrast, NOS2 expression was observed, largely dependent on IFN-gamma signalling. Upon infection with the avirulent PRU strain of T. gondii, IDO mRNA expression was induced in an IFNgammaR-dependent manner. Surprisingly, NOS2 mRNA was severely suppressed. Importantly, we showed in crossing experiments of heterozygote (IFNgammaR+/-) mothers with IFNgammaR-/- males and vice versa that IDO expression largely depends on the presence of IFN-gamma receptors on foetal cells, and to a lesser extent on maternal cells. Immunohistochemical analysis localised foetal IDO production to invasive trophoblasts within the maternal part of the placenta. The placental vascular endothelium only stained positive when the mothers possessed functional IFN-gamma receptors. In contrast, placental NOS2 expression, but also its suppression following infection, seems to be largely dependent on IFN-gamma signalling in maternal cells. Neither factor appears to regulate placental T. gondii growth, as we observed no difference in parasite numbers between (+/-) and (-/-) foetuses. Taken together, our results demonstrate the crucial role of the foetus in placental IDO, but not NOS2, production following T. gondii infection.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Nitric Oxide Synthase Type II/metabolism , Placenta/enzymology , Pregnancy Complications, Parasitic/enzymology , Toxoplasma/physiology , Toxoplasmosis/enzymology , Animals , Female , Fetus/immunology , Fetus/metabolism , Fetus/parasitology , Genes, Protozoan , Immunohistochemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/analysis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Mice, Knockout , Nitric Oxide Synthase Type II/analysis , Nitric Oxide Synthase Type II/genetics , Parasitemia , Placenta/immunology , Placenta/parasitology , Pregnancy , Pregnancy Complications, Parasitic/immunology , RNA, Messenger/analysis , Receptors, Interferon/analysis , Receptors, Interferon/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Toxoplasma/genetics , Toxoplasmosis/immunology , Interferon gamma Receptor
11.
Int Immunopharmacol ; 8(13-14): 1859-62, 2008 Dec 20.
Article in English | MEDLINE | ID: mdl-18809513

ABSTRACT

To elucidate the usefulness of the simultaneous analysis of multiple kinds of soluble cytokine receptors in urine specimens, we determined the levels of both the soluble interferon-gamma receptor alpha chain (sIFN-gammaR1, Th1-type cytokine receptor) and the soluble interleukin 4-receptor alpha chain (sIL-4Ralpha, Th2-type cytokine receptor) in the urine of healthy subjects as reference values and preliminarily applied this method to evaluate patients with diarrhea positive (D+) hemolytic uremic syndrome (HUS) as the diagnostic parameters. The urinary sIFN-gammaR levels of children were significantly lower than those of adults (p < 0.01, n = 107). On the other hand, there was no significant difference between the urine sIL-4R levels of adults and children. Statistical correlation between sIFN-gammaR and sIL-4R values was not observed (p = 0.705). On the day of onset of HUS, the urine sIFN-gammaR levels of the patients (n = 6) with HUS were higher than those of the healthy control group (n = 67) (p < 0.01); however, there was no significant difference in the sIL-4R levels between both groups. The urine evaluation of the balance between the soluble cytokine receptors might be informative for the immune states of HUS patients.


Subject(s)
Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/urine , Receptors, Interferon/analysis , Receptors, Interleukin-4/analysis , Adult , Child , Female , Humans , Male , Urinalysis , Interferon gamma Receptor
12.
Leukemia ; 21(5): 897-905, 2007 May.
Article in English | MEDLINE | ID: mdl-17330098

ABSTRACT

In childhood acute lymphoblastic leukemia (ALL), persistence of leukemic blasts during therapy is of crucial prognostic significance. In the present study, we address molecular and cell biologic features of blasts persisting after 1 week of induction glucocorticoid therapy. Genome-wide gene expression analysis of leukemic samples from precursor B-cell ALL patients (n=18) identified a set of genes differentially expressed in blasts at diagnosis day 0 (d0) and persisting on day 8 (d8). Expression changes indicate a shift towards mature B cells, inhibition of cell cycling and increased expression of adhesion (CD11b/ITGAM) and cytokine (CD119/IFNGR1) receptors. A direct comparison with normal B cells, which are largely therapy resistant, confirmed the differentiation shift at the mRNA (n=10) and protein (n=109) levels. Flow cytometric analysis in independent cohorts of patients confirmed both a decreased proliferative activity (n=13) and the upregulation of CD11b and CD119 (n=29) in d8 blasts. The differentiation shift and low proliferative activity in d8 blasts may account for the persistence of blasts during therapy and affect their sensitivity to further therapeutic treatment. CD11b and CD119 are potential specific markers for d8 blast persistence and detection of minimal residual disease, which warrant further investigation.


Subject(s)
B-Lymphocytes/metabolism , Blast Crisis/metabolism , Gene Expression Profiling , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Adolescent , CD11b Antigen/analysis , Cell Cycle , Cell Proliferation , Child , Child, Preschool , Female , Humans , Infant , Male , Methotrexate/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Prednisone/therapeutic use , Receptors, Interferon/analysis , Interferon gamma Receptor
13.
J Appl Oral Sci ; 26: e20170291, 2018 Oct 04.
Article in English | MEDLINE | ID: mdl-30304122

ABSTRACT

OBJECTIVE: Chronic periodontitis (CP) is characterized by gingival inflammation and bone destruction. It has been reported that interferon-gamma (IFN-γ) levels are high in CP patients; however, the IFN-γ receptor (IFN-γR) has not been studied in gingival tissue from these patients. To evaluate IFN-γ levels and IFN-γR expression in gingival tissue biopsies from chronic periodontitis patients compared with healthy subjects (HS). MATERIAL AND METHODS: Gingival tissues were obtained from all study subjects, CP (n = 18) and healthy subjects (HS) (n = 12). A tissue section of each study subject was embedded in paraffin blocks to determine the expression of IFN-γ R (IFN-γR1 and IFN-γR2) through immunohistochemistry. Another section of the tissue was homogenized and IFN-γ was measured by the ELISA technique. RESULTS: No significant differences were found in the IFN-γR1 expression within the cell layers of the gingival tissue of the study groups. When analyzing the IFN-γR2 expression it was found that IFN-γR2 is strongly expressed in the endothelial cells of CP patients when compared to HS (p<0.05). IFN-γ concentrations in the gingival tissue were significantly higher in CP patients than in HS. No significant correlation between IFN-γ levels and the expression of IFN-γR1 and IFN-γR2 was found. However, a positive correlation between IFN-γ levels and clinical parameters [probing depth (PD) and clinical attachment level (CAL)] was found. CONCLUSION: The study of IFN-γR expression in gingival tissue samples from patients with CP showed an increase only in the IFN-γR2 chain in endothelial cells when compared to HS.


Subject(s)
Chronic Periodontitis/pathology , Endothelial Cells/pathology , Gingiva/pathology , Interferon-gamma/analysis , Receptors, Interferon/analysis , Adult , Biopsy , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Male , Middle Aged , Reference Values , Statistics, Nonparametric , Interferon gamma Receptor
14.
Mol Cell Biol ; 16(5): 2074-82, 1996 May.
Article in English | MEDLINE | ID: mdl-8628273

ABSTRACT

Binding of alpha interferon (IFNalpha) to its receptors induces rapid tyrosine phosphorylation of the receptor subunits IFNaR1 and IFNaR2, the TYK2 and JAK1 tyrosine kinases, and the Stat1 and Stat2 transcription factors. Previous studies have demonstrated that TYK2 directly and specifically binds to and tyrosine phosphorylates IFNaR1 in vitro. We now report a detailed analysis of the TYK2 binding domain on the IFNaR1 subunit. First, we used an in vitro binding assay to identify the TYK2 binding motif in IFNaR1 as well as the critical residues within this region. The most striking feature is the importance of a number of hydrophobic and acidic residues. A minor role is also ascribed to a region resembling the proline-rich "box 1" sequence. In addition, mutations which disrupt in vitro binding also disrupt the coimmunoprecipitation of the receptor and TYK2. We also provide direct evidence that the binding region is both necessary and sufficient to activate TYK2 in vivo. Specifically, mutations in the binding domain act in a dominant-negative fashion to inhibit the IFNalpha-induced tyrosine phosphorylation of TYK2 and Stat2. Further, introduction of dimerized glutathione S-transferase-IFNaR1 fusion proteins into permeabilized cells is sufficient to induce phosphorylation of TYK2 and the receptor, confirming the role of the binding domain in IFNalpha signal transduction. These studies provide clues to the sequences determining the specificity of the association between JAK family tyrosine kinases and cytokine receptors as well as the functional role of these kinases in cytokine signal transduction.


Subject(s)
Interferon-alpha/metabolism , Proteins/metabolism , Receptors, Interferon/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Antibodies , Binding Sites , Cell Line , Glutathione Transferase/biosynthesis , HeLa Cells , Humans , Immunoblotting , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/immunology , Protein-Tyrosine Kinases , Receptor, Interferon alpha-beta , Receptors, Interferon/analysis , Receptors, Interferon/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Spodoptera , TYK2 Kinase , Transfection
15.
Surgery ; 139(6): 743-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16782428

ABSTRACT

OBJECTIVES: Interferons (IFNs) are known to have antiproliferative and immunoregulatory activities that are modulated through specific cellular-surface ligands, known as IFN-alpha, -beta, and -gamma receptors. The presence of these receptors and their impact on survival in patients with pancreatic cancer has not been determined. METHODS: Slides were prepared from 46 patients with pancreatic adenocarcinoma. Immunohistochemistry (IHC) was used subsequently to determine the expression of IFN-alpha/beta receptor-chain 2 (IFNalpha/betaR) and IFN-gamma receptor-chain 1 (IFNgammaR). The correlation among IFN-receptor expression, characteristics of neoplasms, and overall patient survival were determined analytically. RESULTS: The IHC performed for pancreatic adenocarcinoma demonstrated a high IFNalpha/betaR expression in 4% (2/46) of patients, moderate expression in 20% (9/46), and faint or no expression in 76% (35/46). IHC confirmed a high expression of IFNgammaR in 52% (24/46) of patients, moderate expression in 35% (16/46), and faint or no expression in the remaining 13% (6/46). A clinicopathologic survey failed to demonstrate any significant correlation between IFNalpha/betaR and IFNgammaR expression with regard to size of neoplasm, vascular or perineural invasion, lymph node metastases, or stage of disease. Kaplan-Meier survival analyses demonstrated a survival advantage in those patients whose neoplasms expressed moderate to high IFNalpha/betaR expression, compared with those with faint or no IFNalpha/betaR expression (22 vs 13 months; P = .012, log-rank test). The expression of IFNgammaR, however, had no impact on patient survival (20 months vs 17 months; P = .66, log-rank test). CONCLUSIONS: The IFNalpha/betaR is an independent prognostic factor in patients with pancreatic cancer.


Subject(s)
Adenocarcinoma/chemistry , Pancreatic Neoplasms/chemistry , Receptors, Interferon/analysis , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Aged , Female , Humans , Immunohistochemistry , Male , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis
16.
Oncol Rep ; 16(2): 249-55, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16820899

ABSTRACT

Interferon (IFN) is used in the treatment of many malignancies and viral disorders. We recently reported a significant correlation between the efficacy of IFN-alpha combined with chemotherapy in the treatment of advanced hepatocellular carcinoma (HCC) and IFN-alpha/type I IFN receptor (IFNAR2) expression. It is possible that the expression of IFNAR2 in gastrointestinal cancerous tissue, apart from HCC, may predict the efficacy of IFN-alpha combination therapy. We investigated the expression of IFNAR2 in 100 gastrointestinal cancerous tissues. IFNAR2 expression was examined using immunohistochemistry, in surgically resected tissue samples (20 esophageal, 20 gastric, 20 colorectal, 20 cholangiocarcinoma, and 20 pancreatic samples). The expression rate of IFNAR2 was 35.0% (7/20), 25.0% (5/20), 20.0% (4/20), 45.0% (9/20), and 25.0% (5/20) in esophageal, gastric cancer, colorectal, cholangiocarcinoma and pancreatic cancer samples, respectively. In our previous report, the expression rate of IFNAR2 in HCC samples was 64.8% (59/91). Thus, the expression rates of IFNAR2 in the five types of gastrointestinal cancers tested here were low, compared with HCC. The clinical efficacy of IFN-alpha mono- or combination therapies in patients with gastrointestinal neoplasms is expected to be lower than in patients with HCC based on the expression level of IFNAR2.


Subject(s)
Gastrointestinal Neoplasms/drug therapy , Interferon-alpha/therapeutic use , Membrane Proteins/analysis , Receptors, Interferon/analysis , Gastrointestinal Neoplasms/pathology , Humans , Immunohistochemistry , Prognosis , Receptor, Interferon alpha-beta , Treatment Outcome
17.
Cardiovasc Res ; 68(2): 278-88, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16018993

ABSTRACT

BACKGROUND: In a heart with myocarditis, there are cardiomyocytes, inflammatory cells, and non-inflammatory interstitial cells. Immunological molecules are thought to influence not only inflammatory cells but also cardiac function and remodeling. Whatever their origin, the cells they target and the intercellular crosstalk they mediate remain unclear. Here, we examined native gene expression of immunological molecules in normal and rat experimental autoimmune myocarditis (EAM) 18 and 90 days after immunization, using real time RT-PCR in cardiomyocytes, CD11b(+) cells, alphabetaT cells and non-cardiomyocytic non-inflammatory (NCNI) cells. METHODS AND RESULTS: Cells were isolated by collagenase perfusion on a Langendorff apparatus and purified by passing through a stainless-steel sieve followed by magnetic bead column separation using appropriate monoclonal antibodies. Most immunological molecules were expressed in inflammatory cells. However, some were expressed in NCNI cells or cardiomyocytes. Interestingly, most of interleukin (IL)-10, monocyte chemoattractant protein (MCP)-1, or tumor necrosis factor (TNF)-alpha receptor were found in NCNI cells and most of fractalkine were found in NCNI cells and cardiomyocytes. Moreover, TNF-alpha significantly upregulated fractalkine and MCP-1 mRNA in cultivated cells from EAM hearts. CONCLUSION: In the rat experimental myocarditis heart, inflammatory cells express many immunological molecules. Some of them are thought to influence NCNI cells or cardiomyocytes directly via receptors on these cell types. It is further suggested that fractalkine, IL-10, and MCP-1 expressed in NCNI cells or cardiomyocytes regulate inflammatory cells.


Subject(s)
Autoimmune Diseases/immunology , Chemokines/analysis , Myocarditis/immunology , Myocytes, Cardiac/immunology , T-Lymphocytes/immunology , Animals , Chemokine CCL2/analysis , Chemokine CX3CL1 , Chemokines, CX3C/analysis , Fibroblasts/immunology , Flow Cytometry , Hyaluronan Receptors/analysis , Immunohistochemistry/methods , Interleukin-10/analysis , Membrane Proteins/analysis , Muscle, Smooth, Vascular/immunology , Myocardium/immunology , Myocardium/pathology , Osteopontin , Rats , Rats, Inbred Lew , Receptors, Interferon/analysis , Receptors, Tumor Necrosis Factor/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sialoglycoproteins/analysis , Interferon gamma Receptor
19.
Int J Oncol ; 26(1): 129-40, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15586233

ABSTRACT

Based on our previous studies where we found that IFNAR2-1, the short IFNalpha/beta receptor variant, was expressed in pleomorphic sarcoma cells, we decided to determine the relative levels of expression of IFNAR2.1 versus the longer form, named IFNAR2.2, in different pleomorphic sarcoma cells in relation to their response to interferon alpha treatment. When examining a panel of PS cells isolated from surgical specimens, we found that IFNAR2.1 prevailed in 6 out 7 lines analysed and that these generally showed cell cycle arrest and low levels of apoptosis upon IFNalpha treatment. The reverse ratio, i.e. higher constitutive levels of IFNAR2.2 than IFNAR2.1, was associated with an irreversible inhibition of cell growth and pronounced apoptosis. Impairment of tumour growth by low- and high-dose IFNalpha treatment of nude mice inoculated with PS cells expressing predominantly IFNAR2.1 further asserted the effect of the cytokine also in vivo. A proteomic analysis of 120 signalling components in growth arrested, apoptotic PS cells harbouring higher levels of IFNAR2.2 revealed engagement of the canonical Jak/Stat/ISGF3-pathway, the activation of the mitochodrial apoptotic pathway and a potentially novel mechanism of cell cycle blockade unrelated to down-regulation of cyclin A/B and their interacting/regulating kinases. Our results confirm the dominant negative role of IFNAR2.1, but also suggest that the relative endogenous levels of the two IFNalpha/beta receptor isoforms may dictate the signalling pathways triggered by the ligand, such as to cause exclusively cell cycle arrest or induce programmed cell death. This parameter may be of importance for the clinical outcome of IFNalpha treatment of PS.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Interferon-alpha/pharmacology , Membrane Proteins/metabolism , Receptors, Interferon/metabolism , Sarcoma/metabolism , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Membrane/immunology , Cell Proliferation/drug effects , Humans , Male , Membrane Proteins/analysis , Membrane Proteins/genetics , Mice , Neoplasm Transplantation , Protein Isoforms/analysis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteomics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Receptor, Interferon alpha-beta , Receptors, Interferon/analysis , Receptors, Interferon/genetics , Signal Transduction
20.
Fertil Steril ; 83(1): 163-70, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15652903

ABSTRACT

OBJECTIVE: To investigate the expression of the type I interferon receptor (IFNAR) and interferon-induced Mx protein (Mx) in normal human endometrium throughout the menstrual cycle. DESIGN: Prospective study. SETTING: Medical university in Japan. PATIENT(S): Thirty-seven normal endometrial tissues from fertile women who had undergone hysterectomies for reasons other than endometrial disease. INTERVENTION(S): IFNAR-1, IFNAR-2, MxA, and MxB gene expression was analyzed by reverse transcription-real-time quantitative polymerase chain reaction. Moreover, localization of IFNAR-1 and IFNAR-2 were studied by immunohistochemistry. MAIN OUTCOME MEASURE(S): Expression of IFNAR-1, IFNAR-2, MxA, and MxB. RESULT(S): Expression of IFNAR-2 gene was significantly increased in the menstrual and midsecretory phase as compared with in the proliferative phase. Immunohistochemistry for IFNAR-1 and IFNAR-2 revealed weak staining of glandular epithelium and weak staining of stromal cells during the proliferative phase. However, an intense immunohistochemical staining of IFNAR-2 was observed on the surface and basement membrane of glands in the secretory phase. There was no statistical difference between MxA and MxB gene expression throughout the menstrual cycle. CONCLUSION(S): Our results suggest that IFNAR and Mx are expressed in the human endometrium and that the expression of IFNAR is cyclically changed during the menstrual cycle.


Subject(s)
Endometrium/metabolism , GTP-Binding Proteins/genetics , Menstrual Cycle/metabolism , Receptors, Interferon/genetics , Female , Humans , Immunohistochemistry , Myxovirus Resistance Proteins , Prospective Studies , RNA, Messenger/analysis , Receptor, Interferon alpha-beta , Receptors, Interferon/analysis
SELECTION OF CITATIONS
SEARCH DETAIL