Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters










Publication year range
1.
Biomed Res Int ; 2021: 1124055, 2021.
Article in English | MEDLINE | ID: mdl-34812407

ABSTRACT

Autoimmune disorder is a chronic immune imbalance which is developed through a series of pathways. The defect in B cells, T cells, and lack of self-tolerance has been greatly associated with the onset of many types of autoimmune complications including rheumatoid arthritis, systemic lupus erythematosus (SLE), multiple sclerosis, and chronic inflammatory demyelinating polyneuropathy. The SLE is an autoimmune disease with a common type of lupus that causes tissue and organ damage due to the wide spread of inflammation. In the current study, twenty anti-inflammatory peptides derived from plant and animal sources were docked as ligands or peptides counter to proinflammatory cytokines. Interferon gamma (IFN-γ), interleukin 3 (IL-3), and tumor necrosis factor alpha (TNF-α) were targeted in this study as these are involved in the pathogenesis of SLE in many clinical studies. Two docking approaches (i.e., protein-ligand docking and peptide-protein docking) were employed in this study using Molecular Operating Environment (MOE) software and HADDOCK web server, respectively. Amongst docked twenty peptides, the peptide DEDTQAMMPFR with S-score of -11.3018 and HADDOCK score of -10.3 ± 2.5 kcal/mol showed the best binding interactions and energy validation with active amino acids of IFN-γ protein in both docking approaches. Depending upon these results, this peptide could be used as a potential drug candidate to target IFN-γ, IL-3, and TNF-α proteins to control inflammatory events. Other peptides (i.e., QEPQESQQ and FRDEHKK) also revealed good binding affinity with IFN-γ with S-scores of -10.98 and -10.55, respectively. Similarly, the peptides KHDRGDEF, FRDEHKK, and QEPQESQQ showed best binding interactions with IL-3 with S-scores of -8.81, -8.64, and -8.17, respectively.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Interferon-gamma/antagonists & inhibitors , Interleukin-3/antagonists & inhibitors , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Oligopeptides/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Amino Acid Sequence , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/isolation & purification , Drug Discovery/methods , Humans , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Oligopeptides/chemistry , Oligopeptides/genetics , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/pharmacology
2.
Arterioscler Thromb Vasc Biol ; 39(7): 1275-1287, 2019 07.
Article in English | MEDLINE | ID: mdl-31092009

ABSTRACT

Growth factors, such as CSFs (colony-stimulating factors), EGFs (epidermal growth factors), and FGFs (fibroblast growth factors), are signaling proteins that control a wide range of cellular functions. Although growth factor networks are critical for intercellular communication and tissue homeostasis, their abnormal production or regulation occurs in various pathologies. Clinical strategies that target growth factors or their receptors are used to treat a variety of conditions but have yet to be adopted for cardiovascular disease. In this review, we focus on M-CSF (macrophage-CSF), GM-CSF (granulocyte-M-CSF), IL (interleukin)-3, EGFR (epidermal growth factor receptor), and FGF21 (fibroblast growth factor 21). We first discuss the efficacy of targeting these growth factors in other disease contexts (ie, inflammatory/autoimmune diseases, cancer, or metabolic disorders) and then consider arguments for or against targeting them to treat cardiovascular disease. Visual Overview- An online visual overview is available for this article.


Subject(s)
Cardiovascular Diseases/drug therapy , ErbB Receptors/antagonists & inhibitors , Fibroblast Growth Factors/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Humans , Immunotherapy , Interleukin-3/antagonists & inhibitors , Macrophage Colony-Stimulating Factor/antagonists & inhibitors
3.
Immunity ; 50(4): 796-811, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995500

ABSTRACT

The ß common chain cytokines GM-CSF, IL-3, and IL-5 regulate varied inflammatory responses that promote the rapid clearance of pathogens but also contribute to pathology in chronic inflammation. Therapeutic interventions manipulating these cytokines are approved for use in some cancers as well as allergic and autoimmune disease, and others show promising early clinical activity. These approaches are based on our understanding of the inflammatory roles of these cytokines; however, GM-CSF also participates in the resolution of inflammation, and IL-3 and IL-5 may also have such properties. Here, we review the functions of the ß common cytokines in health and disease. We discuss preclinical and clinical data, highlighting the potential inherent in targeting these cytokine pathways, the limitations, and the important gaps in understanding of the basic biology of this cytokine family.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Inflammation/immunology , Interleukin-3/immunology , Interleukin-5/immunology , Animals , Autoimmune Diseases/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Hematopoiesis/immunology , Humans , Inflammation/therapy , Interleukin-3/antagonists & inhibitors , Interleukin-3/deficiency , Interleukin-3/genetics , Interleukin-5/antagonists & inhibitors , Interleukin-5/deficiency , Interleukin-5/genetics , Mice , Mice, Knockout , Multigene Family , Neoplasms/immunology , Neoplasms/therapy , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/immunology , Receptors, Interleukin-5/genetics , Receptors, Interleukin-5/immunology , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Signal Transduction , Structure-Activity Relationship , Vaccination , Wound Healing/immunology
4.
Cell Death Differ ; 25(1): 204-216, 2018 01.
Article in English | MEDLINE | ID: mdl-28960207

ABSTRACT

Basophil granulocytes and mast cells are recognized for their roles in immunity and are central effectors of diverse immunological disorders. Despite their similarities, there is emerging evidence for non-redundant roles of the circulating yet scarce basophils and tissue-resident mast cells, respectively. Because of their importance in allergic pathogenesis, specific induction of apoptosis in basophils and mast cells may represent an interesting novel treatment strategy. The pro-inflammatory cytokine interleukin-3 serves as a key factor for basophil and mouse mast cell survival. Interleukin-3 increases the expression of anti-apoptotic BCL-2 family members, such as BCL-2, BCL-XL or MCL-1; however, little is known how strongly these individual proteins contribute to basophil survival. Here, we were applying small molecule inhibitors called BH3 mimetics, some of which show remarkable success in cancer treatments, to neutralize the function of anti-apoptotic BCL-2 family members. We observed that expression levels of anti-apoptotic BCL-2 proteins do not necessarily correlate with their respective importance for basophil survival. Whereas naive in vitro-differentiated mouse basophils efficiently died upon BCL-2 or BCL-XL inhibition, interleukin-3 priming rendered the cells highly resistant toward apoptosis, and this could only be overcome upon combined targeting of BCL-2 and BCL-XL. Of note, human basophils differed from mouse basophils as they depended on BCL-2 and MCL-1, but not on BCL-XL, for their survival at steady state. On the other hand, and in contrast to mouse basophils, MCL-1 proved critical in mediating survival of interleukin-3 stimulated mouse mast cells, whereas BCL-XL seemed dispensable. Taken together, our results indicate that by choosing the right combination of BH3 mimetic compounds, basophils and mast cells can be efficiently killed, even after stimulation with potent pro-survival cytokines such as interleukin-3. Because of the tolerable side effects of BH3 mimetics, targeting basophils or mast cells for apoptosis opens interesting possibilities for novel treatment approaches.


Subject(s)
Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis , Basophils/metabolism , Mast Cells/metabolism , Aniline Compounds/pharmacology , Animals , Basophils/cytology , Basophils/drug effects , Basophils/enzymology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Caspase 3/metabolism , Cell Survival , Humans , Interleukin-3/antagonists & inhibitors , Interleukin-3/pharmacology , Mast Cells/cytology , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Myeloid Cell Leukemia Sequence 1 Protein/physiology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/physiology , Sulfonamides/pharmacology , bcl-X Protein/antagonists & inhibitors
5.
JCI Insight ; 1(16): e87157, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27734026

ABSTRACT

Little is known about the role of IL-3 in multiple sclerosis (MS) in humans and in experimental autoimmune encephalomyelitis (EAE). Using myelin oligodendrocyte glycoprotein (MOG) peptide-induced EAE, we show that CD4+ T cells are the main source of IL-3 and that cerebral IL-3 expression correlates with the influx of T cells into the brain. Blockade of IL-3 with monoclonal antibodies, analysis of IL-3 deficient mice, and adoptive transfer of leukocytes demonstrate that IL-3 plays an important role for development of clinical symptoms of EAE, for migration of leukocytes into the brain, and for cerebral expression of adhesion molecules and chemokines. In contrast, injection of recombinant IL-3 exacerbates EAE symptoms and cerebral inflammation. In patients with relapsing-remitting MS (RRMS), IL-3 expression by T cells is markedly upregulated during episodes of relapse. Our data indicate that IL-3 plays an important role in EAE and may represent a new target for treatment of MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-3/immunology , Multiple Sclerosis/immunology , Adoptive Transfer , Adult , Animals , Antibodies, Monoclonal , CD4-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Female , Humans , Interleukin-3/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein
6.
Biol Chem ; 397(11): 1187-1204, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27341558

ABSTRACT

The JAK/STAT pathway is an essential mediator of cytokine signaling, often upregulated in human diseases and therefore recognized as a relevant therapeutic target. We previously identified the synthetic chalcone α-bromo-2',3,4,4'-tetramethoxychalcone (α-Br-TMC) as a novel JAK2/STAT5 inhibitor. We also found that treatment with α-Br-TMC resulted in a downward shift of STAT5 proteins in SDS-PAGE, suggesting a post-translational modification that might affect STAT5 function. In the present study, we show that a single cysteine within STAT5 is responsible for the α-Br-TMC-induced protein shift, and that this modification does not alter STAT5 transcriptional activity. We also compared the inhibitory activity of α-Br-TMC to that of another synthetic chalcone, α-trifluoromethyl-2',3,4,4'-tetramethoxychalcone (α-CF3-TMC). We found that, like α-Br-TMC, α-CF3-TMC inhibits JAK2 and STAT5 phosphorylation in response to interleukin-3, however without altering STAT5 mobility in SDS-PAGE. Moreover, we demonstrate that both α-Br-TMC and α-CF3-TMC inhibit interferon-α-induced activation of STAT1 and STAT2, by inhibiting their phosphorylation and the expression of downstream interferon-stimulated genes. Together with the previous finding that α-Br-TMC and α-CF3-TMC inhibit the response to inflammation by inducing Nrf2 and blocking NF-κB activities, our data suggest that synthetic chalcones might be useful as anti-inflammatory, anti-cancer and immunomodulatory agents in the treatment of human diseases.


Subject(s)
Chalcones/pharmacology , Interferon-alpha/antagonists & inhibitors , Interleukin-3/antagonists & inhibitors , Janus Kinase 2/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Amino Acid Sequence , Animals , Cell Line , Humans , Mice , Phosphorylation/drug effects , STAT5 Transcription Factor/chemistry
7.
MAbs ; 8(3): 436-53, 2016.
Article in English | MEDLINE | ID: mdl-26651396

ABSTRACT

The ß common-signaling cytokines interleukin (IL)-3, granulocyte-macrophage colony stimulating factor (GM-CSF) and IL-5 stimulate pro-inflammatory activities of haematopoietic cells via a receptor complex incorporating cytokine-specific α and shared ß common (ßc, CD131) receptor. Evidence from animal models and recent clinical trials demonstrate that these cytokines are critical mediators of the pathogenesis of inflammatory airway disease such as asthma. However, no therapeutic agents, other than steroids, that specifically and effectively target inflammation mediated by all 3 of these cytokines exist. We employed phage display technology to identify and optimize a novel, human monoclonal antibody (CSL311) that binds to a unique epitope that is specific to the cytokine-binding site of the human ßc receptor. The binding epitope of CSL311 on the ßc receptor was defined by X-ray crystallography and site-directed mutagenesis. CSL311 has picomolar binding affinity for the human ßc receptor, and at therapeutic concentrations is a highly potent antagonist of the combined activities of IL-3, GM-CSF and IL-5 on primary eosinophil survival in vitro. Importantly, CSL311 inhibited the survival of inflammatory cells present in induced sputum from human allergic asthmatic subjects undergoing allergen bronchoprovocation. Due to its high potency and ability to simultaneously suppress the activity of all 3 ß common cytokines, CSL311 may provide a new strategy for the treatment of chronic inflammatory diseases where the human ßc receptor is central to pathogenesis. The coordinates for the ßc/CSL311 Fab complex structure have been deposited with the RCSB Protein Data Bank (PDB 5DWU).


Subject(s)
Antibodies, Monoclonal, Murine-Derived , Cytokine Receptor Common beta Subunit , Epitopes , Granulocyte-Macrophage Colony-Stimulating Factor , Interleukin-3 , Interleukin-5 , Animals , Antibodies, Monoclonal, Murine-Derived/chemistry , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/immunology , Asthma/drug therapy , Asthma/immunology , Asthma/pathology , Crystallography, X-Ray , Cytokine Receptor Common beta Subunit/chemistry , Cytokine Receptor Common beta Subunit/immunology , Eosinophils/immunology , Eosinophils/pathology , Epitopes/chemistry , Epitopes/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-3/antagonists & inhibitors , Interleukin-3/immunology , Interleukin-5/antagonists & inhibitors , Interleukin-5/immunology , Mice
8.
Kidney Int ; 88(5): 1088-98, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26131743

ABSTRACT

MRL/lpr mice develop a spontaneous autoimmune disease that closely resembles human systemic lupus erythematosus (SLE) with DNA autoantibodies, hypergammaglobulinemia, immune complex glomerulonephritis, and systemic vasculitis. Little is known about the role of IL-3 in SLE. In order to study this we analyzed the expression of IL-3 in murine lupus and determined whether blockade of IL-3 with a monoclonal antibody or injection of recombinant IL-3 affects lupus nephritis in MRL/lpr mice. During disease progression IL-3 levels were increased in the plasma and in the supernatant of cultured splenocytes from MRL/lpr mice. Administration of IL-3 aggravated the disease with significantly higher renal activity scores, more renal fibrosis, and more glomerular leukocyte infiltration and IgG deposition. Blockade of IL-3 significantly improved acute and chronic kidney damage, reduced the glomerular infiltration of leukocytes and the glomerular deposition of IgG, and decreased the development of renal fibrosis. Furthermore, DNA autoantibody production, proteinuria, and serum creatinine levels were significantly lower in the anti-IL-3 group. Thus, IL-3 plays an important role in the pathogenesis of SLE and the progression of lupus nephritis. Hence, blockade of IL-3 may represent a new strategy for treatment of lupus nephritis.


Subject(s)
Antibodies/pharmacology , Interleukin-3/blood , Interleukin-3/immunology , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Animals , Antibodies, Antinuclear/blood , Cells, Cultured , Creatinine/blood , Disease Progression , Fibrosis , Immunoglobulin G/analysis , Interleukin-3/antagonists & inhibitors , Interleukin-3/pharmacology , Kidney Glomerulus/chemistry , Kidney Glomerulus/pathology , Lupus Nephritis/blood , Lymphocyte Count , Mice , Proteinuria/etiology , Severity of Illness Index , Spleen/cytology
9.
Sci Rep ; 5: 8190, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25644994

ABSTRACT

Chronic Myeloid Leukemia (CML) represents a paradigm for the wider cancer field. Despite the fact that tyrosine kinase inhibitors have established targeted molecular therapy in CML, patients often face the risk of developing drug resistance, caused by mutations and/or activation of alternative cellular pathways. To optimize drug development, one needs to systematically test all possible combinations of drug targets within the genetic network that regulates the disease. The BioModelAnalyzer (BMA) is a user-friendly computational tool that allows us to do exactly that. We used BMA to build a CML network-model composed of 54 nodes linked by 104 interactions that encapsulates experimental data collected from 160 publications. While previous studies were limited by their focus on a single pathway or cellular process, our executable model allowed us to probe dynamic interactions between multiple pathways and cellular outcomes, suggest new combinatorial therapeutic targets, and highlight previously unexplored sensitivities to Interleukin-3.


Subject(s)
Computational Biology/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Algorithms , Apoptosis/drug effects , Computer Simulation , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/metabolism , Gene Knockout Techniques , Gene Regulatory Networks , Humans , Imatinib Mesylate/pharmacology , Interleukin-3/antagonists & inhibitors , Interleukin-3/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Models, Biological , bcl-X Protein/genetics , bcl-X Protein/metabolism , ras Proteins/genetics , ras Proteins/metabolism
10.
Blood ; 122(19): 3335-9, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24041577

ABSTRACT

Chronic myeloid leukemia (CML) stem cells are not dependent on BCR-ABL kinase for their survival, suggesting that kinase-independent mechanisms must contribute to their persistence. We observed that CML stem/progenitor cells (SPCs) produce tumor necrosis factor-α (TNF-α) in a kinase-independent fashion and at higher levels relative to their normal counterparts. We therefore investigated the role of TNF-α and found that it supports survival of CML SPCs by promoting nuclear factor κB/p65 pathway activity and expression of the interleukin 3 and granulocyte/macrophage-colony stimulating factor common ß-chain receptor. Furthermore, we demonstrate that in CML SPCs, inhibition of autocrine TNF-α signaling via a small-molecule TNF-α inhibitor induces apoptosis. Moreover TNF-α inhibition combined with nilotinib induces significantly more apoptosis relative to either treatment alone and a reduction in the absolute number of primitive quiescent CML stem cells. These results highlight a novel survival mechanism of CML SPCs and suggest a new putative therapeutic target for their eradication.


Subject(s)
Chromones/pharmacology , Indoles/pharmacology , Neoplastic Stem Cells/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Leukemic , Humans , Interleukin-3/antagonists & inhibitors , Interleukin-3/genetics , Interleukin-3/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/immunology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptors, Interleukin-3/antagonists & inhibitors , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
11.
Hautarzt ; 61(8): 668-75, 2010 Aug.
Article in German | MEDLINE | ID: mdl-20585746

ABSTRACT

Biologics have been available in Germany for the treatment of moderate to severe chronic plaque psoriasis and/or psoriatic arthritis since 2004. They include chimeric (human/mouse) or fully human monoclonal antibodies or recombinant fusion proteins. The currently available biologics are cytokine antagonists, which neutralize either TNF-alpha or the interleukins IL-12 and IL-23. Unexpected adverse events result either from their potential immunogenicity or from their mode of action, which consists in neutralizing the biologic activity of the respective cytokines. In particular the increased risk for severe infections that may take an atypical course during biologic therapy deserves attention in daily clinical practice.


Subject(s)
Arthritis, Psoriatic/drug therapy , Biological Products/adverse effects , Dermatologic Agents/adverse effects , Drugs, Investigational/adverse effects , Psoriasis/drug therapy , Biological Products/therapeutic use , Dermatologic Agents/therapeutic use , Drugs, Investigational/therapeutic use , Humans , Interleukin-2/antagonists & inhibitors , Interleukin-3/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors
12.
Genome ; 52(5): 409-18, 2009 May.
Article in English | MEDLINE | ID: mdl-19448721

ABSTRACT

Fusarium head blight, predominantly caused by Fusarium graminearum (Schwabe) in North America, is a destructive disease that poses a serious threat to wheat (Triticum aestivum L.) production around the world. cDNA microarrays consisting of wheat ESTs derived from a wheat - F. graminearum interaction suppressive subtractive hybridization library were used to investigate QTL-specific differential gene expression between the resistant Chinese cultivar Sumai-3 and two susceptible near isogenic lines (NILs) following inoculation with F. graminearum. Stringent conditions were employed to reduce the false discovery rate. A total of 25 wheat unigenes were found to express differentially in response to F. graminearum infection. Genes encoding pathogenesis-related (PR) proteins such as beta-1,3-glucanase (PR-2), wheatwins (PR-4), and thaumatin-like proteins (PR-5) showed a significant upregulation in genotypes having the Sumai-3 3BS region. For these three genes, the gene activity was significantly less in the genotype (NIL-3) lacking the Sumai-3 3BS segment. Significant upregulation of phenylalanine ammonia-lyase was detected only in the resistant Sumai-3, indicating the importance of both the 2AL and 3BS regions in the activation of effective defense responses to infection by F. graminearum. Differences in gene expression between the resistant Sumai-3 and the susceptible NILs were found to be mainly quantitative in nature.


Subject(s)
Fusarium/physiology , Genes, Plant , Oligonucleotide Array Sequence Analysis , Plant Diseases/genetics , Quantitative Trait Loci , Triticum/genetics , Biomarkers/metabolism , Blotting, Northern , Gene Expression Profiling , Interleukin-3/antagonists & inhibitors , Interleukin-3/metabolism , Plant Diseases/microbiology , RNA, Neoplasm/metabolism , Triticum/microbiology
13.
Free Radic Biol Med ; 46(2): 244-52, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19013234

ABSTRACT

In the human acute myeloid leukemia cell line M07e, the growth factor interleukin-3 (IL-3) induces ROS formation, positively affecting Glut1-mediated glucose uptake and cell survival. The effect of IL-3 and exogenous hydrogen peroxide on cell viability seems to be mediated through inhibition of the cell death commitment, as shown by apoptotic markers such as caspase activities, apoptotic nuclei, and changes in the amount of proteins belonging to the Bcl-2 family. The pivotal role of ROS is confirmed using various antioxidants, such as EUK-134, ebselen, TEMPO, and hydroxylamine probe. In fact, these antioxidants, acting through different mechanisms, decrease glucose transport activity and cell proliferation activated by IL-3 or by low concentrations of hydrogen peroxide. Moreover, antioxidants foster programmed cell death commitment, as shown by the cited apoptotic parameters. EUK-134, a combined superoxide dismutase/catalase mimetic, opposes the effects of IL-3 and H(2)O(2), decreasing phosphorylation levels of signaling enzymes such as Akt, Src tyrosine kinase, and ERK. Results show that ROS production induced by IL-3 can protect leukemic cells from apoptosis, the effect being counteracted by antioxidants. This mechanism may play an important role in supporting acute myeloid leukemia treatment, thus representing a novel therapeutic strategy.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Glucose Transporter Type 1/metabolism , Leukemia/enzymology , Organometallic Compounds/pharmacology , Salicylates/pharmacology , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin Assembly and Disassembly , Colony-Forming Units Assay , Glucose Transporter Type 1/genetics , Humans , Hydrogen Peroxide/antagonists & inhibitors , Interleukin-3/antagonists & inhibitors , Leukemia/genetics , Leukemia/pathology , Phosphorylation/drug effects , Reactive Oxygen Species/antagonists & inhibitors , Superoxide Dismutase/metabolism , Transfection , Transgenes/genetics
14.
Exp Hematol ; 36(9): 1110-20, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18495327

ABSTRACT

OBJECTIVE: FLRF (Rnf41) gene was identified through screening of subtracted cDNA libraries form murine hematopoietic stem cells and progenitors. Subsequent work has revealed that FLRF acts as E3 ubiquitin ligase, and that it regulates steady-state levels of neuregulin receptor ErbB3 and participates in degradation of IAP protein BRUCE and parkin. The objective of this study was to start exploring the role of FLRF during hematopoiesis. MATERIALS AND METHODS: FLRF was overexpressed in a murine multipotent hematopoietic progenitor cell line EML, which can differentiate into almost all blood cell lineages, and in pro-B progenitor cell line BaF3. The impact of FLRF overexpression on EML cell differentiation into myeloerythroid lineages was studied using hematopoietic colony-forming assays. The interaction of FLRF with cytokine receptors and receptor levels in control cells and EML and BaF3 cells overexpressing FLRF were examined with Western and immunoprecipitation. RESULTS: Remarkably, overexpression of FLRF significantly attenuated erythroid and myeloid differentiation of EML cells in response to cytokines erythropoietin (EPO) and interleukin-3 (IL-3), and retinoic acid (RA), and resulted in significant and constitutive decrease of steady-state levels of IL-3, EPO, and RA receptor-alpha (RARalpha) in EML and BaF3 cells. Immunoprecipitation has revealed that FLRF interacts with IL-3, EPO, and RARalpha receptors in EML and BaF3 cells, and that FLRF-mediated downregulation of these receptors is ligand binding-independent. CONCLUSIONS: The results of this study have revealed new FLRF-mediated pathway for ligand-independent receptor level regulation, and support the notion that through maintaining basal levels of cytokine receptors, FLRF is involved in the control of hematopoietic progenitor cell differentiation into myeloerythroid lineages.


Subject(s)
Carrier Proteins/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/drug effects , Receptors, Erythropoietin/biosynthesis , Receptors, Interleukin-3/biosynthesis , Receptors, Retinoic Acid/physiology , Animals , Cell Line/drug effects , Cell Line/metabolism , Cell Lineage , Colony-Forming Units Assay , Down-Regulation/physiology , Erythropoietin/antagonists & inhibitors , Erythropoietin/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/metabolism , Interleukin-3/antagonists & inhibitors , Interleukin-3/pharmacology , Ligands , Mice , Protein Binding , Receptors, Erythropoietin/genetics , Receptors, Interleukin-3/genetics , Receptors, Retinoic Acid/genetics , Recombinant Fusion Proteins/physiology , Retinoic Acid Receptor alpha , Tretinoin/antagonists & inhibitors , Tretinoin/pharmacology , Ubiquitin-Protein Ligases
15.
Int Immunopharmacol ; 7(5): 656-61, 2007 May.
Article in English | MEDLINE | ID: mdl-17386413

ABSTRACT

IMUNOR, a low-molecular weight (< 12 kD) ultrafiltered pig leukocyte extract, has been previously found to have significant stimulatory effects on murine hematopoiesis supressed by ionizing radiation or cytotoxic drugs. This communication shows data on the mechanisms of these effects. Using ELISA assay, significantly increased levels of granulocyte colony-stimulating factor (G-CSF) and interleukin-6 (IL-6) were observed. On the contrary, no detectable levels of granulocyte-macrophage colony-stimulating factor (GM-CFC) and interleukin-3 (IL-3) have been found in blood serum of IMUNOR-treated mice. Incubation of the serum from IMUNOR-treated mice with antibodies against G-CSF caused abrogation of the ability of the sera to stimulate in vitro growth of colonies originating from granulocyte-macrophage progenitor cells (GM-CFC). In contrast, incubation of the serum with antibodies against IL-6 did not change its colony-stimulating activity. It may be inferred from these findings that G-CSF is probably the main cytokine responsible for the granulopoiesis-stimulating effects of IMUNOR. When the serum from IMUNOR-treated mice with G-CSF inactivated by anti-G-CSF antibodies (but with elevated IL-6) was added to cultures of bone marrow cells together with a suboptimum concentration of IL-3, a significant increase in the numbers of GM-CFC colonies was found. Moreover, conjoint inactivation of G-CSF and IL-6 significantly decreased the numbers of GM-CFC colonies in comparison with those observed when only G-CSF was inactivated. This observation strongly suggests that though IMUNOR-induced IL-6 is not able to induce the growth of GM-CFC colonies alone, it is able to potentiate the hematopoiesis-stimulating effect of IL-3. These findings represent a new knowledge concerning the hematopoiesis-stimulating action of IMUNOR, a promising immunomodulatory agent.


Subject(s)
Granulocyte Colony-Stimulating Factor/physiology , Granulocytes/physiology , Hematopoiesis/drug effects , Immune Sera/pharmacology , Interleukin-6/physiology , Leukocytes/physiology , Tissue Extracts/pharmacology , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Extracts , Enzyme-Linked Immunosorbent Assay , Female , Granulocyte Colony-Stimulating Factor/antagonists & inhibitors , Interleukin-3/antagonists & inhibitors , Interleukin-6/antagonists & inhibitors , Mice , Stimulation, Chemical , Swine
16.
J Immunol ; 177(1): 77-83, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16785501

ABSTRACT

Eosinophils are involved in a variety of allergic, parasitic, malignant, and idiopathic disorders by releasing a variety of factors including specific granule proteins, lipid mediators, and proinflammatory and immunoregulatory cytokines and chemokines. In addition, they interact with various cell types in the inflamed tissue. Yet, the mechanism of eosinophil activation is still poorly understood. Recently, we described the expression and function of the CD2-subfamily of receptors and especially 2B4 on human eosinophils. In this study we focus on CD48, the high-affinity ligand of 2B4. CD48 is a GPI-anchored protein involved in cellular activation, costimulation, and adhesion, but has not been studied on eosinophils. We demonstrate that human eosinophils from atopic asthmatics display enhanced levels of CD48 expression and that IL-3 up-regulates CD48 expression. Furthermore, cross-linking CD48 on human eosinophils triggers release of eosinophil granule proteins. Assessment of CD48 expression in a murine model of experimental asthma revealed that CD48 is induced by allergen challenge and partially regulated by IL-3. Additionally, anti-IL-3 reduces CD48 expression and the degree of airway inflammation. Thus, CD48 is an IL-3-induced activating receptor on eosinophils, likely involved in promoting allergic inflammation.


Subject(s)
Allergens/physiology , Antigens, CD/physiology , Eosinophils/immunology , Eosinophils/metabolism , Interleukin-3/physiology , Allergens/administration & dosage , Animals , Antigens, CD/biosynthesis , Antigens, CD/blood , Asthma/immunology , Asthma/metabolism , Asthma/pathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , CD48 Antigen , Cells, Cultured , Disease Models, Animal , Eosinophil Peroxidase/metabolism , Eosinophils/enzymology , Female , Humans , Interleukin-3/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Mice, Transgenic , Nasal Polyps/immunology , Nasal Polyps/pathology , Peritonitis/immunology , Peritonitis/metabolism , Peritonitis/pathology , Up-Regulation/immunology
17.
Anticancer Res ; 26(2A): 905-16, 2006.
Article in English | MEDLINE | ID: mdl-16619486

ABSTRACT

Cytokines interact with cell-surface receptors, initiating signaling cascades that promote cell growth while inhibiting the pathways of apoptotic cells. Rin1 is a multifunctional protein that has been shown to regulate EGF receptor signaling and endocytosis. To examine the role of Rin1 in IL3 receptor signaling pathways, Rin1 and deletion mutants were expressed in cells using a retrovirus system. In this study, the overexpression of Rin1 molecules was shown to selectively block IL-3 activation of the Ras-Erk1/2 and PI3K/Akt pathways and the IL-3-stimulated incorporation of [3H] thymidine into DNA without a significant effect on the activity of the JNK and p38K pathways. Moreover, the depletion of Rin1 by RNA interference induced cell growth. In addition, Rin1 was also required as a downstream effector of BCR/ABL-induced cell proliferation. Interestingly, the expression of Rin1 selectively blocked the activation of Erk1/2 induced by the BCR/ABL oncogene. These results demonstrate that Rin1 plays an essential and selective role in both IL3- and BCR/ABL-induced cell proliferation and highlight a new function for Rin1 in leukemic cells.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Receptors, Interleukin-3/metabolism , Cell Growth Processes/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fusion Proteins, bcr-abl/metabolism , Gene Deletion , HL-60 Cells , Humans , Interleukin-3/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , K562 Cells , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , ras Proteins/metabolism
18.
Cell Signal ; 18(2): 174-82, 2006 Feb.
Article in English | MEDLINE | ID: mdl-15982852

ABSTRACT

Hematopoietic cytokines, including interleukin (IL)-3 and erythropoietin (Epo), regulate hematopoiesis by stimulating their receptors coupled with the Jak2 tyrosine kinase to induce receptor tyrosine phosphorylation and activate mainly the STAT5, PI3K/Akt, and Ras/MEK/ERK signaling pathways. Here we demonstrate that IL-3 or Epo induces a rapid and transient (peaking at 30 min) as well as late progressive increase in reactive oxygen species (ROS) in a hematopoietic progenitor model cell line, 32Dcl3, and its subclone expressing the Epo receptor (EpoR), 32D/EpoR-Wt. The cytokine-induced ROS generation was not affected in 32Dcl3 cells depleted of mitochondrial DNA. The antioxidant N-acetyl-L-cysteine (NAC) inhibited IL-3-induced tyrosine phosphorylation of Jak2, IL-3 receptor betac subunit (IL-3Rbetac), and STAT5 as well as activation-specific phosphorylation of Akt, MEK, and ERK, while treatment of cells with H2O2 activated these signaling events. NAC also inhibited the EpoR-induced transphosphorylation of IL-3Rbetac. Moreover, NAC treatment reduced the expression levels of c-Myc, Cyclin D2, and Cyclin E, and induced expression of p27, thus inhibiting the G1 to S phase transition of cells cultured with IL-3. Further studies have shown that the degradation of c-Myc was facilitated or inhibited by treatment of cells with NAC or H2O2, respectively. These data indicate that the rapid generation of ROS by cytokine stimulation, which is at least partly independent of mitochondria, may play a role in activation of Jak2 and the STAT5, PI3K/Akt, and Ras/MEK/ERK signaling pathways as well as in transactivation of cytokine receptors. The cytokine-induced ROS generation was also implicated in G1 to S progression, possibly through stabilization of c-Myc and induction of G1 phase Cyclin expression leading to suppression of p27.


Subject(s)
Erythropoietin/pharmacology , Hematopoietic Stem Cells/metabolism , Interleukin-3/pharmacology , Reactive Oxygen Species/metabolism , Receptors, Erythropoietin/metabolism , Receptors, Interleukin-3/metabolism , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line , Erythropoietin/antagonists & inhibitors , Hematopoietic Stem Cells/drug effects , Interleukin-3/antagonists & inhibitors , Mice , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction
19.
Cytokine ; 30(5): 248-53, 2005 Jun 07.
Article in English | MEDLINE | ID: mdl-15927849

ABSTRACT

The nature of erythropoietin (EPO)-dependent, erythroid cell regulatory factors secreted by endothelial cells is largely unknown. The production of thrombospondin 1 (TSP-1) and insulin-like growth factor binding protein 3 (IGFBP-3) is increased in cultures of human umbilical vein endothelial cells (HUVEC) incubated with erythropoietin (EPO). Simultaneous incubation of HUVEC with EPO and interleukin 3 (IL-3) resulted in a decreased production, suggesting that both TSP-1 and IGFBP-3 belong to the EPO- and IL-3-dependent erythroid regulatory factors previously described in cultures of bone marrow endothelial cells. TSP-1 and TSP-1 derived synthetic peptides based on the CD36 and CD47 binding sites of TSPs increased thymidine incorporation into bovine erythroid cells of fetal liver. IGBBP-3 inhibited thymidine incorporation in the same cells. Preincubation of erythroid cells with TSP-1 eliminated the inhibitory activity of IGFBP-3. We suggest that EPO-dependent, endothelial-derived TSP-1 may play a positive role in red cell production by acting directly on erythroid cells, stimulating DNA synthesis and preventing the inhibitory action of IGFBP-3.


Subject(s)
Endothelial Cells/metabolism , Erythroid Cells/metabolism , Erythropoietin/pharmacology , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Thrombospondin 1/metabolism , Thymidine/metabolism , Umbilical Cord/metabolism , Animals , Cattle , Cells, Cultured , Endothelial Cells/drug effects , Erythroid Cells/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 3/metabolism , Interleukin-3/antagonists & inhibitors , Interleukin-3/metabolism , Peptide Fragments/pharmacology , Thrombospondin 1/pharmacology , Umbilical Cord/drug effects
20.
J Immunol ; 173(9): 5721-9, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15494524

ABSTRACT

The anti-inflammatory effects of salicylates, originally attributed to inhibition of cyclooxygenase activity, are currently known to involve additional mechanisms. In this study we investigated the possible modulation by salicylates of NFAT-mediated transcription in lymphocytic and monocytic cell lines. RNase protection assays showed that 2-acetoxy-4-trifluoromethylbenzoic acid (triflusal) inhibited, in a dose-dependent manner, mRNA expression of several cytokine genes, most of which are NFAT-regulated and cyclosporin A (CsA)-sensitive. In Jurkat cells, the expression of IL-3, GM-CSF, TNF-alpha, TGF-beta1, IL-2, lymphotactin, MIP-1alpha, and MIP-1beta was inhibited to different extents. In THP-1 cells, inhibition of the expression of M-CSF, G-CSF, stem cell factor, IFN-gamma, TNF-alpha, TGF-beta1, lymphotoxin-beta1, MIP-1alpha, MIP-1beta, and IL-8 was observed. Sodium salicylate and aspirin only showed significant effects at 5 mM. The transcriptional activity of two genes that contain NFAT sites, a GM-CSF full promoter and a T cell-specific enhancer from the IL-3 locus, was also inhibited by salicylates. Transactivation experiments performed with several NFAT-dependent and AP-1-dependent reporter genes showed that triflusal strongly inhibited NFAT-dependent transcription at concentrations as low as 0.25 mM. Sodium salicylate and aspirin were less potent. The triflusal inhibitory effect was reversible and synergized with suboptimal doses of CsA. Experiments to address the mechanism of action of salicylates in the NFAT activation cascade disclosed a mechanism different from that of CsA, because salicylates inhibited DNA-binding and NFAT-mediated transactivation without affecting phosphorylation or subcellular localization of NFAT. In summary, these data describe a new pharmacological effect of salicylates as inhibitors of NFAT-dependent transcription.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Gene Silencing/drug effects , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Salicylates/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription, Genetic/drug effects , Animals , Cell Line, Transformed , Cell Line, Tumor , Cells, Cultured , Chemokines/antagonists & inhibitors , Chemokines/biosynthesis , Chemokines/genetics , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Cytokines/genetics , DNA-Binding Proteins/physiology , Down-Regulation/drug effects , Down-Regulation/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Gene Silencing/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Interleukin-3/antagonists & inhibitors , Interleukin-3/genetics , Jurkat Cells , Mice , NFATC Transcription Factors , Nuclear Proteins/physiology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/immunology , Transcription Factors/physiology , Transcription, Genetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...