Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
J Immunol ; 205(10): 2629-2639, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33067379

ABSTRACT

The Bruton's tyrosine kinase (BTK) inhibitor ibrutinib has profound activity in chronic lymphocytic leukemia (CLL) but limited curative potential by itself. Residual signaling pathways that maintain survival of CLL cells might be targeted to improve ibrutinib's therapeutic activity, but the nature of these pathways is unclear. Ongoing activation of IFN receptors in patients on ibrutinib was suggested by the presence of type I and II IFN in blood together with the cycling behavior of IFN-stimulated gene (ISG) products when IFN signaling was blocked intermittently with the JAK inhibitor ruxolitinib. IFN signaling in CLL cells from human patients was not prevented by ibrutinib in vitro or in vivo, but ISG expression was significantly attenuated in vitro. ISGs such as CXCL10 that require concomitant activation of NF-κB were decreased when this pathway was inhibited by ibrutinib. Other ISGs, exemplified by LAG3, were decreased as a result of inhibited protein translation. Effects of IFN on survival remained intact as type I and II IFN-protected CLL cells from ibrutinib in vitro, which could be prevented by ruxolitinib and IFNR blocking Abs. These observations suggest that IFNs may help CLL cells persist and specific targeting of IFN signaling might deepen clinical responses of patients on ibrutinib.


Subject(s)
Adenine/analogs & derivatives , Interferon Type I/metabolism , Interferon-gamma/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Piperidines/pharmacology , Adenine/pharmacology , Adenine/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/immunology , Cell Survival/drug effects , Cell Survival/immunology , Drug Resistance, Neoplasm/immunology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Nitriles , Piperidines/therapeutic use , Primary Cell Culture , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines , Receptors, Interferon/antagonists & inhibitors , Receptors, Interferon/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Cells, Cultured
2.
PLoS Pathog ; 14(8): e1007246, 2018 08.
Article in English | MEDLINE | ID: mdl-30142226

ABSTRACT

Chronic activation of the immune system in HIV infection is one of the strongest predictors of morbidity and mortality. As such, approaches that reduce immune activation have received considerable interest. Previously, we demonstrated that administration of a type I interferon receptor antagonist (IFN-1ant) during acute SIV infection of rhesus macaques results in increased virus replication and accelerated disease progression. Here, we administered a long half-life PASylated IFN-1ant to ART-treated and ART-naïve macaques during chronic SIV infection and measured expression of interferon stimulated genes (ISG) by RNA sequencing, plasma viremia, plasma cytokines, T cell activation and exhaustion as well as cell-associated virus in CD4 T cell subsets sorted from peripheral blood and lymph nodes. Our study shows that IFN-1ant administration in both ART-suppressed and ART-untreated chronically SIV-infected animals successfully results in reduction of IFN-I-mediated inflammation as defined by reduced expression of ISGs but had no effect on plasma levels of IL-1ß, IL-1ra, IL-6 and IL-8. Unlike in acute SIV infection, we observed no significant increase in plasma viremia up to 25 weeks after IFN-1ant administration or up to 15 weeks after ART interruption. Likewise, cell-associated virus measured by SIV gag DNA copies was similar between IFN-1ant and placebo groups. In addition, evaluation of T cell activation and exhaustion by surface expression of CD38, HLA-DR, Ki67, LAG-3, PD-1 and TIGIT, as well as transcriptome analysis showed no effect of IFN-I blockade. Thus, our data show that blocking IFN-I signaling during chronic SIV infection suppresses IFN-I-related inflammatory pathways without increasing virus replication, and thus may constitute a safe therapeutic intervention in chronic HIV infection.


Subject(s)
Anti-Retroviral Agents/pharmacology , Inflammation/prevention & control , Interferon Type I/antagonists & inhibitors , Simian Acquired Immunodeficiency Syndrome , T-Lymphocytes/drug effects , Virus Replication/drug effects , Animals , Anti-Retroviral Agents/therapeutic use , Chronic Disease , Inflammation/immunology , Inflammation/virology , Interferon Type I/metabolism , Lymphocyte Activation/drug effects , Macaca mulatta , Receptors, Interferon/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/immunology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , T-Lymphocytes/immunology
3.
Pak J Pharm Sci ; 33(1(Supplementary)): 393-401, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32122873

ABSTRACT

Breast cancer is the most common cancer in females, and the leading cause of cancer-related mortality in the world. Among the available treatment options for cancer, chemotherapy is the therapy for treating a variety of cancer patients. However, the therapeutic efficacy of current agents is minimal and these drugs do not retard the progression of disease pathology. Lack of appropriate therapy may increase the prevalence of disease in world. Hence, more effective strategies and novel therapies must be pursued for altering the progression of the disease acting through different mechanisms. There is a continuing need for new and improved therapy. Hence, Vitamin B17 is suggested a therapeutic potential for treating breast cancer. This study is to evaluate the potential therapy of vitamin B17 (Vit B17, amygdalin) against Ehrlish solid tumors, bearing mice (EST) induced DNA damage, NF-Kb, TNFα and apoptosis. Sixty female mice were randomly divided into four groups: (I, control group; II, VitB17 group; III, EST group; IV, EST+VitB17 group). EST induced group had elevated in the levels of serum ALT, AST, ALP, creatinine, urea, potassium ions, cholesterol, triglycerides, cytokine IFNγ, NF-kb, DNA damage, tumor TNF-α, VEGF expressions and had an associated reduction in serum albumin, total proteins, sodium ions, tumor NF-kb, Bcl2 and survivin expressions. Treatment of EST with vitamin B17 (EST+VitB17) modulates the changes in liver and kidney functions, electrolytes, cytokines, NF-kb and apoptosis in mice bearing EST. Hence, these findings suggest that vitamin B17 can be a reliable and novel therapy for breast cancer, further validate the neoplastic activity of Vitamin B17 as a potential therapy for other types of cancer is needed.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/metabolism , DNA Fragmentation/drug effects , Vitamin B Complex/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Ehrlich Tumor/pathology , Female , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Interferon/antagonists & inhibitors , Receptors, Interferon/metabolism , Survivin/antagonists & inhibitors , Survivin/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vitamin B Complex/pharmacology , Interferon gamma Receptor
4.
J Immunol ; 193(10): 5099-107, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25297874

ABSTRACT

Listeria monocytogenes is a major cause of mortality resulting from food poisoning in the United States. In mice, C5 has been genetically linked to host resistance to listeriosis. Despite this genetic association, it remains poorly understood how C5 and its activation products, C5a and C5b, confer host protection to this Gram-positive intracellular bacterium. In this article, we show in a systemic infection model that the major receptor for C5a, C5aR1, is required for a normal robust host immune response against L. monocytogenes. In comparison with wild-type mice, C5aR1(-/-) mice had reduced survival and increased bacterial burden in their livers and spleens. Infected C5aR1(-/-) mice exhibited a dramatic reduction in all major subsets of splenocytes, which was associated with elevated caspase-3 activity and increased TUNEL staining. Because type 1 IFN has been reported to impede the host response to L. monocytogenes through the promotion of splenocyte death, we examined the effect of C5aR1 on type 1 IFN expression in vivo. Indeed, serum levels of IFN-α and IFN-ß were significantly elevated in L. monocytogenes-infected C5aR1(-/-) mice. Similarly, the expression of TRAIL, a type 1 IFN target gene and a proapoptotic factor, was elevated in NK cells isolated from infected C5aR1(-/-) mice. Treatment of C5aR1(-/-) mice with a type 1 IFNR blocking Ab resulted in near-complete rescue of L. monocytogenes-induced mortality. Thus, these findings reveal a critical role for C5aR1 in host defense against L. monocytogenes through the suppression of type 1 IFN expression.


Subject(s)
Interferon-alpha/genetics , Interferon-beta/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Spleen/immunology , Anaphylatoxins/immunology , Animals , Antibodies/pharmacology , Apoptosis , Bacterial Load , Caspase 3/genetics , Caspase 3/immunology , Complement C5a/genetics , Complement C5a/immunology , Complement C5b/genetics , Complement C5b/immunology , Gene Expression , Interferon-alpha/immunology , Interferon-beta/immunology , Listeriosis/drug therapy , Listeriosis/microbiology , Listeriosis/mortality , Liver/immunology , Liver/microbiology , Liver/pathology , Lymphocytes/immunology , Lymphocytes/microbiology , Lymphocytes/pathology , Male , Mice , Mice, Knockout , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/immunology , Receptors, Interferon/antagonists & inhibitors , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Spleen/microbiology , Spleen/pathology , Survival Analysis , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/immunology
5.
PLoS Pathog ; 7(11): e1002345, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22072965

ABSTRACT

Migratory lung dendritic cells (DCs) transport viral antigen from the lungs to the draining mediastinal lymph nodes (MLNs) during influenza virus infection to initiate the adaptive immune response. Two major migratory DC subsets, CD103(+) DCs and CD11b(high) DCs participate in this function and it is not clear if these antigen presenting cell (APC) populations become directly infected and if so whether their activity is influenced by the infection. In these experiments we show that both subpopulations can become infected and migrate to the draining MLN but a difference in their response to type I interferon (I-IFN) signaling dictates the capacity of the virus to replicate. CD103(+) DCs allow the virus to replicate to significantly higher levels than do the CD11b(high) DCs, and they release infectious virus in the MLNs and when cultured ex-vivo. Virus replication in CD11b(high) DCs is inhibited by I-IFNs, since ablation of the I-IFN receptor (IFNAR) signaling permits virus to replicate vigorously and productively in this subset. Interestingly, CD103(+) DCs are less sensitive to I-IFNs upregulating interferon-induced genes to a lesser extent than CD11b(high) DCs. The attenuated IFNAR signaling by CD103(+) DCs correlates with their described superior antigen presentation capacity for naïve CD8(+) T cells when compared to CD11b(high) DCs. Indeed ablation of IFNAR signaling equalizes the competency of the antigen presenting function for the two subpopulations. Thus, antigen presentation by lung DCs is proportional to virus replication and this is tightly constrained by I-IFN. The "interferon-resistant" CD103(+) DCs may have evolved to ensure the presentation of viral antigens to T cells in I-IFN rich environments. Conversely, this trait may be exploitable by viral pathogens as a mechanism for systemic dissemination.


Subject(s)
Dendritic Cells/immunology , Influenza A Virus, H1N1 Subtype/immunology , Interferon Type I/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Adaptive Immunity , Animals , Antigen Presentation , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/virology , Antigens, CD/analysis , Antigens, Viral/immunology , CD11b Antigen/analysis , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Dendritic Cells/metabolism , Dendritic Cells/virology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/physiology , Integrin alpha Chains/analysis , Interferon Type I/metabolism , Lymph Nodes/immunology , Lymph Nodes/virology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/virology , Receptors, Interferon/antagonists & inhibitors , Virus Replication
6.
Nat Genet ; 6(1): 90-7, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8136841

ABSTRACT

Gene targeting in somatic cells represents a potentially powerful method for gene therapy, yet with the exception of pluripotent mouse embryonic stem (ES) cells, homologous recombination has not been reported for a well characterized, non-transformed mammalian cell. Applying a highly efficient strategy for targeting an integral membrane protein--the interferon gamma receptor--in ES cells, we have used homologous recombination to target a non-transformed somatic cell, the mouse myoblast, and to compare targeting efficiencies in these two cell types. Gene-targeted myoblasts display the properties of normal cells including normal morphology, ability to differentiate in vitro, stable diploid karyotype, inability to form colonies in soft agar and lack of tumorigenicity in nude mice.


Subject(s)
Genetic Therapy/methods , Muscles/immunology , Receptors, Interferon/genetics , Animals , Base Sequence , Genetic Techniques , Genetic Vectors , Major Histocompatibility Complex , Mice , Mice, Nude , Molecular Sequence Data , Muscles/cytology , Receptors, Interferon/antagonists & inhibitors , Recombination, Genetic , Stem Cells/cytology , Stem Cells/immunology , Interferon gamma Receptor
7.
Infect Immun ; 80(9): 3039-48, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22689821

ABSTRACT

Mycobacterium avium subsp. paratuberculosis is the causative agent of Johne's disease in cattle and may have implications for human health. Establishment of chronic infection by M. avium subsp. paratuberculosis depends on its subversion of host immune responses. This includes blocking the ability of infected macrophages to be activated by gamma interferon (IFN-γ) for clearance of this intracellular pathogen. To define the mechanism by which M. avium subsp. paratuberculosis subverts this critical host cell function, patterns of signal transduction to IFN-γ stimulation of uninfected and M. avium subsp. paratuberculosis-infected bovine monocytes were determined through bovine-specific peptide arrays for kinome analysis. Pathway analysis of the kinome data indicated activation of the JAK-STAT pathway, a hallmark of IFN-γ signaling, in uninfected monocytes. In contrast, IFN-γ stimulation of M. avium subsp. paratuberculosis-infected monocytes failed to induce patterns of peptide phosphorylation consistent with JAK-STAT activation. The inability of IFN-γ to induce differential phosphorylation of peptides corresponding to early JAK-STAT intermediates in infected monocytes indicates that M. avium subsp. paratuberculosis blocks responsiveness at, or near, the IFN-γ receptor. Consistent with this hypothesis, increased expression of negative regulators of the IFN-γ receptors SOCS1 and SOCS3 as well as decreased expression of IFN-γ receptor chains 1 and 2 is observed in M. avium subsp. paratuberculosis-infected monocytes. These patterns of expression are functionally consistent with the kinome data and offer a mechanistic explanation for this critical M. avium subsp. paratuberculosis behavior. Understanding this mechanism may contribute to the rational design of more effective vaccines and/or therapeutics for Johne's disease.


Subject(s)
Interferon-gamma/antagonists & inhibitors , Monocytes/immunology , Monocytes/microbiology , Mycobacterium avium subsp. paratuberculosis/pathogenicity , Paratuberculosis/immunology , Paratuberculosis/microbiology , Receptors, Interferon/antagonists & inhibitors , Animals , Cattle , Cattle Diseases/immunology , Cattle Diseases/microbiology , Interferon-gamma/immunology , Mycobacterium avium subsp. paratuberculosis/immunology , Paratuberculosis/pathology , Phosphorylation , Protein Processing, Post-Translational , Receptors, Interferon/immunology , Signal Transduction
8.
Circulation ; 119(10): 1424-32, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19255340

ABSTRACT

BACKGROUND: Atherosclerosis is an inflammatory disease in which interferon (IFN)-gamma, the signature cytokine of Th1 cells, plays a central role. We investigated whether interleukin (IL)-17, the signature cytokine of Th17 cells, is also associated with human coronary atherosclerosis. METHODS AND RESULTS: Circulating IL-17 and IFN-gamma were detected in a subset of patients with coronary atherosclerosis and in referent outpatients of similar age without cardiac disease but not in young healthy individuals. IL-17 plasma levels correlated closely with those of the IL-12/IFN-gamma/CXCL10 cytokine axis but not with known Th17 inducers such as IL-1beta, IL-6, and IL-23. Both IL-17 and IFN-gamma were produced at higher levels by T cells within cultured atherosclerotic coronary arteries after polyclonal activation than within nondiseased vessels. Combinations of proinflammatory cytokines induced IFN-gamma but not IL-17 secretion. Blockade of IFN-gamma signaling increased IL-17 synthesis, whereas neutralization of IL-17 responses decreased IFN-gamma synthesis; production of both cytokines was inhibited by transforming growth factor-beta1. Approximately 10-fold fewer coronary artery-infiltrating T helper cells were IL-17 producers than IFN-gamma producers, and unexpectedly, IL-17/IFN-gamma double producers were readily detectable within the artery wall. Although IL-17 did not modulate the growth or survival of cultured vascular smooth muscle cells, IL-17 interacted cooperatively with IFN-gamma to enhance IL-6, CXCL8, and CXCL10 secretion. CONCLUSIONS: Our findings demonstrate that IL-17 is produced concomitantly with IFN-gamma by coronary artery-infiltrating T cells and that these cytokines act synergistically to induce proinflammatory responses in vascular smooth muscle cells.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Coronary Artery Disease/pathology , Inflammation Mediators/metabolism , Interferon-gamma/physiology , Interleukin-17/physiology , Myocytes, Smooth Muscle/pathology , T-Lymphocyte Subsets/metabolism , Vasculitis/etiology , Adult , Aged , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/metabolism , Coronary Artery Disease/complications , Coronary Artery Disease/immunology , Coronary Vessels/drug effects , Female , Gene Expression Regulation/drug effects , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Interleukin-17/biosynthesis , Interleukin-17/metabolism , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Interleukin-8/biosynthesis , Interleukin-8/metabolism , Interleukins/pharmacology , Male , Middle Aged , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Receptors, Interferon/antagonists & inhibitors , Receptors, Interferon/immunology , Receptors, Interleukin-17/antagonists & inhibitors , Receptors, Interleukin-17/immunology , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology , Vasculitis/physiopathology , Interferon gamma Receptor
9.
Rheumatol Int ; 30(5): 591-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19547978

ABSTRACT

Atherosclerotic cardiovascular disease (ASCVD) contributes to morbidity and mortality in systemic lupus erythematosus (SLE). Immunologic derangements may disrupt cholesterol balance in vessel wall monocytes/macrophages and endothelium. We determined whether lupus plasma impacts expression of cholesterol 27-hydroxylase, an anti-atherogenic cholesterol-degrading enzyme that promotes cellular cholesterol efflux, in THP-1 human monocytes and primary human aortic endothelial cells (HAEC). THP-1 monocytes and HAEC were incubated in medium containing SLE patient plasma or apparently healthy control human plasma (CHP). SLE plasma decreased 27-hydroxylase message in THP-1 monocytes by 47 +/- 8% (p < 0.008) and in HAEC by 51 +/- 5.5% (n = 5, p < 0.001). THP-1 macrophages were incubated in 25% lupus plasma or CHP and cholesterol-loaded (50 microg ml(-1) acetylated low density lipoprotein). Lupus plasma more than doubled macrophage foam cell transformation (74 +/- 3% vs. 35 +/- 3% for CHP, n = 3, p < 0.001). Impaired cholesterol homeostasis in SLE provides further evidence of immune involvement in atherogenesis. Strategies to inhibit or reverse arterial cholesterol accumulation may benefit SLE patients.


Subject(s)
Atherosclerosis/immunology , Autoimmunity , Cholestanetriol 26-Monooxygenase/metabolism , Cholesterol/metabolism , Endothelial Cells/enzymology , Lupus Erythematosus, Systemic/blood , Monocytes/enzymology , Adolescent , Adult , Atherosclerosis/blood , Case-Control Studies , Cells, Cultured , Cholestanetriol 26-Monooxygenase/genetics , Down-Regulation , Endothelial Cells/immunology , Female , Foam Cells/enzymology , Gene Expression Regulation, Enzymologic , Homeostasis , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Lipoproteins, LDL/metabolism , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/immunology , Monocytes/immunology , RNA, Messenger/metabolism , Receptors, Interferon/antagonists & inhibitors , Receptors, Interferon/immunology , Risk Factors , Young Adult , Interferon gamma Receptor
10.
J Leukoc Biol ; 107(2): 273-284, 2020 02.
Article in English | MEDLINE | ID: mdl-31793076

ABSTRACT

Legionella pneumophila is an opportunistic human pathogen and causative agent of the acute pneumonia known as Legionnaire's disease. Upon inhalation, the bacteria replicate in alveolar macrophages (AM), within an intracellular vacuole termed the Legionella-containing vacuole. We recently found that, in vivo, IFNγ was required for optimal clearance of intracellular L. pneumophila by monocyte-derived cells (MC), but the cytokine did not appear to influence clearance by AM. Here, we report that during L. pneumophila lung infection, expression of the IFNγ receptor subunit 1 (IFNGR1) is down-regulated in AM and neutrophils, but not MC, offering a possible explanation for why AM are unable to effectively restrict L. pneumophila replication in vivo. To test this, we used mice that constitutively express IFNGR1 in AM and found that prevention of IFNGR1 down-regulation enhanced the ability of AM to restrict L. pneumophila intracellular replication. IFNGR1 down-regulation was independent of the type IV Dot/Icm secretion system of L. pneumophila indicating that bacterial effector proteins were not involved. In contrast to previous work, we found that signaling via type I IFN receptors was not required for IFNGR1 down-regulation in macrophages but rather that MyD88- or Trif- mediated NF-κB activation was required. This work has uncovered an alternative signaling pathway responsible for IFNGR1 down-regulation in macrophages during bacterial infection.


Subject(s)
Legionella pneumophila/growth & development , Legionnaires' Disease/microbiology , Lung/microbiology , Macrophages, Alveolar/microbiology , NF-kappa B/metabolism , Receptors, Interferon/antagonists & inhibitors , Animals , Down-Regulation , Interferon Type I/metabolism , Legionella pneumophila/metabolism , Legionnaires' Disease/metabolism , Lung/metabolism , Macrophages, Alveolar/metabolism , Mice , Mice, Transgenic , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Signal Transduction , Interferon gamma Receptor
11.
J Immunother Cancer ; 7(1): 152, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31196219

ABSTRACT

BACKGROUND: T-cell mediated immunotherapy brought clinical success for many cancer patients. Nonetheless, downregulation of MHC class I antigen presentation, frequently occurring in solid cancers, limits the efficacy of these therapies. Unraveling the mechanisms underlying this type of immune escape is therefore of great importance. We here investigated the immunological effects of metabolic stress in cancer cells as a result of nutrient deprivation. METHODS: TC1 and B16F10 tumor cell lines were cultured under oxygen- and glucose-deprivation conditions that mimicked the tumor microenvironment of solid tumors. Presentation of peptide antigens by MHC class I molecules was measured by flow cytometry and via activation of tumor-specific CD8 T cell clones. The proficiency of the IFNy-STAT1 pathway was investigated by Western blots on phosphorylated proteins, transfection of constitutive active STAT1 constructs and qPCR of downstream targets. Kinase inhibitors for PI3K were used to examine its role in IFNy receptor signal transduction. RESULTS: Combination of oxygen- and glucose-deprivation resulted in decreased presentation of MHC class I antigens on cancer cells, even in the presence of the stimulatory cytokine IFNy. This unresponsiveness to IFNy was the result of failure to phosphorylate the signal transducer STAT1. Forced expression of constitutive active STAT1 fully rescued the MHC class I presentation. Furthermore, oxygen- and glucose-deprivation increased PI3K activity in tumor cells. Pharmacological inhibition of this pathway not only restored signal transduction through IFNy-STAT1 but also improved MHC class I presentation. Importantly, PI3K inhibitors also rendered tumor cells sensitive for recognition by CD8 T cells in culture conditions of metabolic stress. CONCLUSIONS: These data revealed a strong impact of metabolic stress on the presentation of tumor antigens by MHC class I and suggest that this type of tumor escape takes place at hypoxic areas even during times of active T cell immunity and IFNy release.


Subject(s)
Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Interferon/antagonists & inhibitors , Animals , Antigen Presentation , Cell Hypoxia , Cell Line, Tumor , Glucose/deficiency , Histocompatibility Antigens Class I/immunology , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Mice , Mice, Inbred C57BL , Phosphorylation , Receptors, Interferon/immunology , Receptors, Interferon/metabolism , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , Signal Transduction , Stress, Physiological/immunology , Tumor Escape , Interferon gamma Receptor
12.
Br J Pharmacol ; 176(2): 317-333, 2019 01.
Article in English | MEDLINE | ID: mdl-30338847

ABSTRACT

BACKGROUND AND PURPOSE: Sepsis is a serious clinical condition with a high mortality rate. Anti inflammatory agents have been found to be beneficial for the treatment of sepsis. Here, we have evaluated the anti-inflammatory activity of seselin in models of sepsis and investigated the underlying molecular mechanism(s). EXPERIMENTAL APPROACH: In vivo therapeutic effects of seselin was evaluated in two models of sepsis, caecal ligation and puncture or injection of LPS, in C57BL/6 mice. In vitro, anti-inflammatory activity of seselin was assessed with macrophages stimulated with LPS and IFN-γ. Anti inflammatory actions were analysed with immunohistochemical methods, ELISA and Western blotting. Flow cytometry was used to assess markers of macrophage phenotype (pro- or anti-inflammatory). Other methods used included co-immunoprecipitation, cellular thermal shift assay and molecular docking. KEY RESULTS: In vivo, seselin clearly ameliorated sepsis induced by caecal ligation and puncture. In lung tissue from septic mice and in cultured macrophages, seselin down-regulated levels of proinflammatory factors and activity of STAT1 and p65, the master signal pathway molecules for polarization of macrophages into the proinflammatory phenotype. Importantly, adoptive transfer of bone marrow-derived macrophages, pretreated with seselin, lowered systemic proinflammatory factors in mice challenged with LPS. The underlying mechanism was that seselin targeted Jak2 to block interaction with IFNγ receptors and downstream STAT1. CONCLUSIONS AND IMPLICATIONS: Seselin exhibited anti-inflammatory activity through its action on Jak2. These results indicated a possible application of seselin to the treatment of inflammatory disease via blocking the development of the proinflammatory phenotype of macrophages.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Coumarins/pharmacology , Inflammation/drug therapy , Janus Kinase 2/antagonists & inhibitors , Macrophages/drug effects , Sepsis/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Coumarins/administration & dosage , Disease Models, Animal , Female , Inflammation/metabolism , Janus Kinase 2/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Phenotype , Receptors, Interferon/antagonists & inhibitors , Receptors, Interferon/metabolism , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/metabolism , Sepsis/metabolism
13.
Virology ; 522: 19-26, 2018 09.
Article in English | MEDLINE | ID: mdl-30014854

ABSTRACT

Interferons (IFNs) exhibit forceful inhibitory activities against numerous viruses by inducing synthesis of anti-viral proteins or promoting immune cell functions, which help eradicate the vicious microbes. Consequently, the degree to which viruses evade or counterattack IFN responses influences viral pathogenicity. Viruses have developed many strategies to interfere with the synthesis of IFNs or IFN receptor signaling pathway. Furthermore, multiple viruses decrease levels of IFN receptors via diverse tactics, which include decreasing type I IFN receptor mRNA expression, blocking post-translational modification of the receptor, and degrading IFN receptors. Recently, influenza virus was found to induce CK1α-induced phosphorylation and subsequent degradation of the receptor for type I and II IFNs. In this review, viral mechanisms that remove IFN receptors are summarized with an emphasis on the mechanisms for virus-induced degradation of IFN receptors.


Subject(s)
Host-Pathogen Interactions , Immune Evasion , Receptors, Interferon/antagonists & inhibitors , Viruses/pathogenicity , Animals , Humans
14.
Leukemia ; 32(11): 2483-2494, 2018 11.
Article in English | MEDLINE | ID: mdl-29691471

ABSTRACT

The therapeutic benefits of allogeneic hematopoietic stem cell transplantation (allo-HSCT) are derived from the graft-versus-leukemia (GvL) effects of the procedure. There is a strong association between the GvL effects and graft-versus-host disease (GvHD), a major life-threatening complication of allo-HSCT. The limiting of GvHD while maintaining the GvL effect remains the goal of allo-HSCT. Therefore, identifying optimal therapeutic targets to selectively suppress GvHD while maintaining the GvL effects represents a significant unmet medical need. We demonstrate that the dual inhibition of interferon gamma receptor (IFNγR) and interleukin-6 receptor (IL6R) results in near-complete elimination of GvHD in a fully major histocompatibility complex-mismatched allo-HSCT model. Furthermore, baricitinib (an inhibitor of Janus kinases 1 and 2 (JAK1/JAK2) downstream of IFNγR/IL6R) completely prevented GvHD; expanded regulatory T cells by preserving JAK3-STAT5 signaling; downregulated CXCR3 and helper T cells 1 and 2 while preserving allogeneic antigen-presenting cell-stimulated T-cell proliferation; and suppressed the expression of major histocompatibility complex II (I-Ad), CD80/86, and PD-L1 on host antigen-presenting cells. Baricitinib also reversed established GvHD with 100% survival, thus demonstrating both preventive and therapeutic roles for this compound. Remarkably, baricitinib enhanced the GvL effects, possibly by downregulating tumor PD-L1 expression.


Subject(s)
Azetidines/pharmacology , Graft vs Host Disease/metabolism , Graft vs Host Disease/prevention & control , Receptors, Interferon/antagonists & inhibitors , Receptors, Interleukin-6/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , B7-H1 Antigen/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Hematopoietic Stem Cell Transplantation/methods , Janus Kinase 1/metabolism , Male , Mice , Purines , Pyrazoles , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous/methods , Interferon gamma Receptor
15.
Arch Neurol ; 63(11): 1579-84, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17101826

ABSTRACT

BACKGROUND: Interferon inhibitory activity (IIA) is a logical candidate for explaining neutralizing antibody-negative partial responsiveness to interferon beta in multiple sclerosis (MS), but its role has not been evaluated. OBJECTIVE: To investigate the role of IIA and soluble interferon-alpha/beta receptor (sIFNR) in determining response of patients with MS to interferon beta therapy. DESIGN: Parallel-group, open-label study. SETTING: Baird Multiple Sclerosis Center, Buffalo, NY. Patients Blood was obtained before and 24 hours after injection of interferon beta-1a from 38 anti-interferon beta neutralizing antibody-negative patients with relapsing-remitting MS and 16 untreated healthy controls. On the basis of clinical parameters of response to interferon beta therapy, the patients were divided into stable or good-responder (n = 20) and active or partial-responder (n = 18) groups. MAIN OUTCOME MEASURES: Quantitative analyses of magnetic resonance imaging were obtained; the IIA and sIFNR levels were measured using bioassay and enzyme-linked immunosorbent assay, respectively. RESULTS: The IIA and sIFNR levels were elevated in MS patients compared with controls (P<.001). The IIA levels were higher in active or partial responders compared with stable or good responders (P<.001); the sIFNR levels were not different between groups. The Extended Disability Status Score and T2 lesion volumes were higher in the active or partial-responder group compared with the stable or good-responder group. Interferon beta-1a did not have short-term effects on the IIA and sIFNR levels. In univariate general linear model and stepwise regression analyses, IIA levels were associated with T2 lesion volume. CONCLUSION: The levels of IIA are associated with increased MS disease activity and with responsiveness to interferon beta therapy in anti-interferon beta neutralizing antibody-negative MS patients.


Subject(s)
Interferon-beta/antagonists & inhibitors , Interferon-beta/blood , Multiple Sclerosis/blood , Receptors, Interferon/antagonists & inhibitors , Adult , Antibodies/blood , Case-Control Studies , Cell Line , Disability Evaluation , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Interferon beta-1a , Interferon-beta/immunology , Interferon-beta/therapeutic use , Magnetic Resonance Imaging/methods , Male , Middle Aged , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Receptors, Interferon/blood , beta 2-Microglobulin/blood
16.
Wei Sheng Wu Xue Bao ; 46(5): 802-6, 2006 Oct.
Article in Zh | MEDLINE | ID: mdl-17172032

ABSTRACT

The function of NS4B is incompletely understood. The aim of the study is to understand the influence of NS4B on anti-viral response. After cell line stably expressing NS4B established, the influence of IFN-alpha of different concentration on VSV was studied using plaque assay; cell expression profiling caused by NS4B was studied using DNA microarray, and the IFNGR1 fluorescence intensity was analyzed. Our data showed that HCV-NS4B could suppress immuno-associated gene expression, in particular, IFN-gamma receptor signal transduction-related genes. Taken together, NS4B could play some roles in HCV resistance to IFN therapy.


Subject(s)
Receptors, Interferon/antagonists & inhibitors , Signal Transduction/physiology , Viral Nonstructural Proteins/physiology , Drug Resistance, Viral , Gene Expression Regulation , HeLa Cells , Hepatitis C/drug therapy , Humans , Interferon-gamma/therapeutic use , Oligonucleotide Array Sequence Analysis , Receptors, Interferon/physiology , Interferon gamma Receptor
17.
Leukemia ; 16(6): 1135-42, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12040445

ABSTRACT

Affinity purified, freshly isolated CD34+ progenitors were shown to express low levels of type I interferon (IFN) receptors (740 +/- 60 binding sites/cell, K(d) 0.7 +/- 0.04 nM) determined by Scatchard's analysis using a radiolabelled, neutralizing, monoclonal antibody directed against the IFNAR1 chain of the human type I IFN receptor. Treatment of freshly isolated (day 0), highly purified (>95% pure) CD34+ cells with recombinant IFN-alpha resulted in rapid tyrosine phosphorylation and activation of STAT1, Tyk2 and JAK1 as shown by Western immunoblotting. Similarly, IFN treatment was shown by confocal microscopy to result in rapid nuclear localization of the transcription factors IRF1 and STAT2, demonstrating the presence of functional IFN receptors on freshly isolated (day 0) CD34+ cells. The number of specific type I IFN receptor binding sites expressed on hematopoietic progenitor cells increased to some 1440 +/- 40 per cell after 11 days of cultivation of CD34+ cells in vitrosuggesting that receptor expression increases with cell differentiation. IFN-mediated signal transduction and the inhibitory effect of IFN-alpha on 7 or 14 days CFU-GM and BFU-E colony formation was abrogated in the presence of the anti-IFNAR1 mAb, indicating that IFN-alpha acts directly on the proliferation of human hematopoietic progenitor cells via receptor activated signal transduction without excluding the induction of other cytokines or growth factors by residual accessory cells.


Subject(s)
Antigens, CD34/analysis , Hematopoietic Stem Cells/metabolism , Receptors, Interferon/physiology , Signal Transduction , Active Transport, Cell Nucleus , Antibodies, Monoclonal/pharmacology , Cell Division/drug effects , Cell Nucleus/metabolism , Cells, Cultured , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/chemistry , Humans , Interferon Regulatory Factor-1 , Interferon-alpha/antagonists & inhibitors , Janus Kinase 1 , Kinetics , Membrane Proteins , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Receptor, Interferon alpha-beta , Receptors, Interferon/antagonists & inhibitors , Receptors, Interferon/immunology , STAT1 Transcription Factor , STAT2 Transcription Factor , TYK2 Kinase , Trans-Activators/metabolism
18.
Int Immunopharmacol ; 29(2): 401-407, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26507164

ABSTRACT

Pro-inflammatory cytokines play a crucial role in the etiology of atopic dermatitis. We demonstrated that Herba Epimedii has anti-inflammatory potential in an atopic dermatitis mouse model; however, limited research has been conducted on the anti-inflammatory effects and mechanism of icariin, the major active ingredient in Herba Epimedii, in human keratinocytes. In this study, we evaluated the anti-inflammatory potential and mechanisms of icariin in the tumor necrosis factor-α (TNF-α)/interferon-γ (IFN-γ)-induced inflammatory response in human keratinocytes (HaCaT cells) by observing these cells in the presence or absence of icariin. We measured IL-6, IL-8, IL-1ß, MCP-1 and GRO-α production by ELISA; IL-6, IL-8, IL-1ß, intercellular adhesion molecule-1 (ICAM-1) and tachykinin receptor 1 (TACR1) mRNA expression by real-time PCR; and P38-MAPK, P-ERK and P-JNK signaling expression by western blot in TNF-α/IFN-γ-stimulated HaCaT cells before and after icariin treatment. The expression of TNF-α-R1 and IFN-γ-R1 during the stimulation of the cell models was also evaluated before and after icariin treatment. We investigated the effect of icariin on these pro-inflammatory cytokines and detected whether this effect occurred via the mitogen-activated protein kinase (MAPK) signal transduction pathways. We further specifically inhibited the activity of two kinases with 20µM SB203580 (a p38 kinase inhibitor) and 50µM PD98059 (an ERK1/2 kinase inhibitor) to determine the roles of the two signal pathways involved in the inflammatory response. We found that icariin inhibited TNF-α/IFN-γ-induced IL-6, IL-8, IL-1ß, and MCP-1 production in a dose-dependent manner; meanwhile, the icariin treatment inhibited the gene expression of IL-8, IL-1ß, ICAM-1 and TACR1 in HaCaT cells in a time- and dose-dependent manner. Icariin treatment resulted in a reduced expression of p-P38 and p-ERK signal activation induced by TNF-α/IFN-γ; however, only SB203580, the p38 alpha/beta inhibitor, inhibited the secretion of inflammatory cytokines induced by TNF-α/IFN-γ in cultured HaCaT cells. The differential expression of TNF-α-R1 and IFN-γ-R1 was also observed after the stimulation of TNF-α/IFN-γ, which was significantly normalized after the icariin treatment. Collectively, we illustrated the anti-inflammatory property of icariin in human keratinocytes. These effects were mediated, at least partially, via the inhibition of substance P and the p38-MAPK signaling pathway, as well as by the regulation of the TNF-α-R1 and IFN-γ-R1 signals.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Flavonoids/pharmacology , Inflammation/prevention & control , Interferon-gamma/antagonists & inhibitors , Keratinocytes/drug effects , MAP Kinase Signaling System/drug effects , Substance P/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Cell Line , Cytokines/metabolism , Dose-Response Relationship, Drug , Humans , Inflammation/chemically induced , Receptors, Interferon/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Signal Transduction/drug effects
19.
J Interferon Cytokine Res ; 15(5): 395-402, 1995 May.
Article in English | MEDLINE | ID: mdl-7544231

ABSTRACT

Hemin and other metalloporphyrins are known as very versatile compounds in nature, because they are able to carry out numerous functions in a free state or in association with specific proteins. When Friend murine erythroleukemia cells are treated with IFN-beta plus 100 microM hemin, the antiviral state is not observed, whereas the antiviral effect of IFN-gamma is unaffected by hemin treatment. This inhibitory effect of hemin is not restricted to erythroid cells. In fact, it is also observed in murine L929 and in human cell lines treated with IFN-beta. Neither trivalent iron in other forms nor hemin analogs (such as protoporphyrin IX or Sn(2+)-protoporphyrine IX) mimic this effect. Conversely, Co(3+)-protoporphyrin IX was as effective as hemin. At the transcriptional level, results obtained by run-on assays on nuclei from IFN-treated cells indicate that hemin does not completely inhibit IFN-beta induction of 2-5A synthetase gene(s) at 6 h of treatment but abolishes it at 24 h. In addition, hemin is able to inhibit the accumulation of IFN-induced 2-5A synthetase mRNAs. Experiments carried out to investigate the hemin effect on the early steps of the IFN signaling pathway indicate that hemin interferes with the ability of type I IFN to bind to its receptor, probably by a direct action on the IFN molecule.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Hemin/pharmacology , Interferon-beta/antagonists & inhibitors , Animals , Cell Line , Encephalomyocarditis virus/drug effects , Ferric Compounds/pharmacology , Hemin/analogs & derivatives , Humans , Interferon Type I/antagonists & inhibitors , Interferon Type I/metabolism , Interferon-beta/genetics , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Mice , Protoporphyrins/pharmacology , Quaternary Ammonium Compounds/pharmacology , RNA/biosynthesis , Receptors, Interferon/antagonists & inhibitors , Receptors, Interferon/metabolism , Recombinant Proteins , Transcription, Genetic , Tumor Cells, Cultured , Vesicular stomatitis Indiana virus/drug effects
20.
Arch Immunol Ther Exp (Warsz) ; 45(1): 93-9, 1997.
Article in English | MEDLINE | ID: mdl-9090447

ABSTRACT

Radioiodinated human recombinant interferon alpha 88 (125I-HuIFN-alpha 88) binds to high affinity receptors of IFN sensitive Daudi cells (Burkitt lymphoma cell line). Suramin, a low molecular weight (1429), polyanionic compound at concentrations 105-175 microM completely abolished 125I-IFN-alpha 88 binding to Daudi cells at low temperature (4 degrees C). At 37 degrees C, however, its effect was only partial and depended on incubation time. Suramin also dissociated IFN-alpha 88-receptor complexes but, upon incubation of cells with IFN at 37 degrees C IFN-receptor complexes, became gradually less sensitive to suramin action. Dissociation by suramin IFN-alpha 88-receptor complexes prevented induction of (2-5)A synthetase activity and inhibited down-regulation of IFN receptors on Daudi cells. We suppose that the first reaction which represents IFN binding to the surface receptors is inhibited and dissociated by suramin, but when IFN is transferred to a tight activation complexes on the cell membrane or internalized, such complexes cannot be dissociated by suramin.


Subject(s)
Interferon-alpha/metabolism , Receptors, Interferon/drug effects , Suramin/pharmacology , 2',5'-Oligoadenylate Synthetase/antagonists & inhibitors , 2',5'-Oligoadenylate Synthetase/biosynthesis , Binding, Competitive/immunology , Burkitt Lymphoma , Down-Regulation/drug effects , Enzyme Induction/drug effects , Humans , Iodine Radioisotopes , Protein Binding/drug effects , Protein Binding/immunology , Receptors, Interferon/antagonists & inhibitors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL