Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Oncol Lett ; 14(5): 5497-5504, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29113177

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO)-induced immunosuppression can be clinically beneficial for autoimmune diseases. Primary biliary cirrhosis (PBC) is characterized by autoimmune lesions of intrahepatic bile duct epithelial cells that may lead to irreversible cirrhosis or hepatocellular carcinoma. The present study assessed the expression and function of IDO in a cell culture model and in PBC patients. IDO expression was monitored in a human immortalized but non-malignant biliary epithelial cell (iBEC) line. Increased expression of IDO1/2 was observed in the iBECs following stimulation with interferon-γ (IFN-γ). The induction of IDO was IFN-γ-dependent, but was independent of the transforming growth factor-ß (TGF-ß) pathway. IDO enzymatic activity was observed in the supernatant of iBECs following stimulation with IFN-γ using colorimetric assays. A total of 47 serum samples from PBC patients were used to examine IDO activity by high-performance liquid chromatography, with samples from 24 healthy volunteers used as controls. Patients with PBC exhibited an increased rate of tryptophan to kynurenine conversion (P>0.01). Liver sections from patients with PBC (n=5) and those of healthy controls (n=5) were used for immunohistochemical studies. IDO expression was observed in biliary epithelial cells and in hepatocytes of PBC patients. Finally, the effect of tryptophan metabolites on human cluster of differentiation (CD) 4+ T cells in inducing polarization towards a regulatory T cell phenotype was examined. 3-Hydroxykynurenine significantly upregulated the fraction of CD4+ cells expressing forkhead box p3 (Foxp3). The results of the present study suggest a therapeutic opportunity for the management of PBC and indicate that tryptophan catabolism could serve as a potential biomarker to monitor disease progression.

2.
J Crohns Colitis ; 11(5): 610-620, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28453768

ABSTRACT

BACKGROUND AND AIMS: The αEß7 integrin is crucial for retention of T lymphocytes at mucosal surfaces through its interaction with E-cadherin. Pathogenic or protective functions of these cells during human intestinal inflammation, such as ulcerative colitis [UC], have not previously been defined, with understanding largely derived from animal model data. Defining this phenotype in human samples is important for understanding UC pathogenesis and is of translational importance for therapeutic targeting of αEß7-E-cadherin interactions. METHODS: αEß7+ and αEß7- colonic T cell localization, inflammatory cytokine production and expression of regulatory T cell-associated markers were evaluated in cohorts of control subjects and patients with active UC by immunohistochemistry, flow cytometry and real-time PCR of FACS-purified cell populations. RESULTS: CD4+αEß7+ T lymphocytes from both healthy controls and UC patients had lower expression of regulatory T cell-associated genes, including FOXP3, IL-10, CTLA-4 and ICOS in comparison with CD4+αEß7- T lymphocytes. In UC, CD4+αEß7+ lymphocytes expressed higher levels of IFNγ and TNFα in comparison with CD4+αEß7- lymphocytes. Additionally the CD4+αEß7+ subset was enriched for Th17 cells and the recently described Th17/Th1 subset co-expressing both IL-17A and IFNγ, both of which were found at higher frequencies in UC compared to control. CONCLUSION: αEß7 integrin expression on human colonic CD4+ T cells was associated with increased production of pro-inflammatory Th1, Th17 and Th17/Th1 cytokines, with reduced expression of regulatory T cell-associated markers. These data suggest colonic CD4+αEß7+ T cells are pro-inflammatory and may play a role in UC pathobiology.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Colitis, Ulcerative/immunology , Colon/cytology , Integrins/immunology , Adult , Aged , Case-Control Studies , Colitis, Ulcerative/metabolism , Colon/immunology , Cytokines/metabolism , Female , Flow Cytometry , Humans , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Young Adult
3.
Sci Rep ; 7: 44384, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28290520

ABSTRACT

Chemokines promote leukocyte recruitment during inflammation. The oxidative burst is an important effector mechanism, this leads to the generation of reactive nitrogen species (RNS), including peroxynitrite (ONOO). The current study was performed to determine the potential for nitration to alter the chemical and biological properties of the prototypical CC chemokine, CCL2. Immunofluorescence was performed to assess the presence of RNS in kidney biopsies. Co-localisation was observed between RNS-modified tyrosine residues and the chemokine CCL2 in diseased kidneys. Nitration reduced the potential of CCL2 to stimulate monocyte migration in diffusion gradient chemotaxis assays (p < 0.05). This was consistent with a trend towards reduced affinity of the nitrated chemokine for its cognate receptor CCR2b. The nitrated chemokine was unable to induce transendothelial monocyte migration in vitro and failed to promote leukocyte recruitment when added to murine air pouches (p < 0.05). This could potentially be attributed to reduced glycosaminoglycan binding ability, as surface plasmon resonance spectroscopy showed that nitration reduced heparan sulphate binding by CCL2. Importantly, intravenous administration of nitrated CCL2 also inhibited the normal recruitment of leukocytes to murine air pouches filled with unmodified CCL2. Together these data suggest that nitration of CCL2 during inflammation provides a mechanism to limit and resolve acute inflammation.


Subject(s)
Chemokine CCL2/metabolism , Inflammation/metabolism , Reactive Nitrogen Species/metabolism , Receptors, CCR2/metabolism , Animals , Binding Sites , Cell Movement/genetics , Chemokine CCL2/chemistry , Chemokines/metabolism , Inflammation/pathology , Leukocytes/metabolism , Mice , Monocytes/metabolism , Oxidative Stress/genetics , Peroxynitrous Acid/chemistry , Peroxynitrous Acid/metabolism , Reactive Nitrogen Species/chemistry , Receptors, CCR2/genetics , Tyrosine/metabolism
5.
J Clin Immunol ; 36(2): 117-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26801501

ABSTRACT

PURPOSE: To investigate the clinical and functional aspects of MST1 (STK4) deficiency in a profoundly CD4-lymphopenic kindred with a novel homozygous nonsense mutation in STK4. Although recent studies have described the cellular effects of murine Mst1 deficiency, the phenotype of MST1-deficient human lymphocytes has yet to be fully explored. Patient lymphocytes were therefore investigated in the context of current knowledge of murine Mst1 deficiency. METHODS: Genetic etiology was identified by whole exome sequencing of genomic DNA from two siblings, combined with linkage analysis in the wider family. MST1 protein expression was assessed by immunoblotting. The ability of patient lymphocytes to adhere to ICAM-1 under flow conditions was measured, and transwell assays were used to assess chemotaxis. Chemokine receptor expression was examined by flow cytometry and receptor signalling by immunoblotting. RESULTS: A homozygous nonsense mutation in STK4 (c.442C > T, p.Arg148Stop) was found in the patients, leading to a lack of MST1 protein expression. Patient leukocytes exhibited deficient chemotaxis after stimulation with CXCL11, despite preserved expression of CXCR3. Patient lymphocytes were also unable to bind effectively to immobilised ICAM-1 under flow conditions, in keeping with a failure to develop high affinity binding. CONCLUSION: The observed abnormalities of adhesion and migration imply a profound trafficking defect among human MST1-deficient lymphocytes. By analogy with murine Mst1 deficiency and other defects of leucocyte trafficking, this is likely to contribute to immunodeficiency by impairing key aspects of T-cell development and function such as positive selection in the thymus, thymic egress and immune synapse formation in the periphery.


Subject(s)
Cell Adhesion/genetics , Chemotaxis, Leukocyte/genetics , Genes, Recessive , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Protein Serine-Threonine Kinases/deficiency , Child, Preschool , Female , Humans , Immunologic Deficiency Syndromes/metabolism , Immunophenotyping , Intercellular Adhesion Molecule-1/metabolism , Intracellular Signaling Peptides and Proteins , Lymphocytes/immunology , Lymphocytes/metabolism , Pedigree , Phenotype , Siblings
6.
Immunology ; 143(2): 138-45, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24912917

ABSTRACT

Oxidative stress is a major and recurring cause of damage during inflammation, especially following organ transplantation. Initial ischaemia-reperfusion injury causes the production of many reactive oxygen and nitrogen species, and subsequent recruitment and activation of inflammatory cells can lead to further oxidative stress. This stress is well known to cause damage at the cellular level, for example by induction of senescence leading to the production of a characteristic senescence-associated secretory phenotype. Chemokines are an important component of the senescence-associated secretory phenotype, recruiting further leucocytes and reinforcing the stress and senescence responses. As well as inducing the production of proteins, including chemokines, oxidative stress can alter proteins themselves, both directly and by induction of enzymes capable of modification. These alterations can lead to important modifications to their biological activity and also alter detection by some antibodies, potentially limiting the biological relevance of some immunochemical and proteomic biomarkers. Peroxynitrite, a reactive nitrogen species generated during inflammation and ischaemia, can cause such modifications by nitrating chemokines. Matrix metalloproteinases, released by many stressed cells, can cleave chemokines, altering function, while peptidylarginine deiminases can inactivate certain chemokines by citrullination. This review discusses the relationship between inflammation and post-translational modification, focusing on the functional modulation of transplant-relevant pro-inflammatory chemokines.


Subject(s)
Chemokines/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Organ Transplantation/adverse effects , Animals , Biomarkers/metabolism , Humans , Inflammation/immunology , Oxidative Stress , Protein Processing, Post-Translational , Signal Transduction , Treatment Outcome
8.
Transplantation ; 94(5): 456-64, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22890131

ABSTRACT

BACKGROUND: Regulatory T cells (Tregs) effectively ameliorate graft-versus-host disease (GVHD). The mechanisms underlying Treg therapeutic effect on GVHD are not fully elucidated. This study investigates whether Treg prevention of GVH tissue damage is associated with blocking CD8 effector T-cell tissue invasion, a question not yet addressed in humans. METHOD: Tissue-infiltrating T cells and histopathology scores were detected using an in vitro human GVHD skin explant model, together with immunohistochemistry, cytometric bead array, functional adhesion and migration assays, flow cytometry, and quantitative real-time polymerase chain reaction. RESULTS: Treg intervention during priming significantly decreased effector T-cell infiltration into target tissue (P<0.01) resulting in a striking reduction in the histopathology score of tissue injury (P<0.0001). These results were coupled with reduced CXCR3 and cutaneous lymphocyte antigen expression by effector T cells, together with decreased CXCL10 and CXCL11 expression in target tissue. Treg intervention also impaired the functional interaction of CXCR3 and cutaneous lymphocyte antigen with their specific ligands (P<0.01) and suppressed the secretion of CXCL9, CXCL10, and interferon-γ (P<0.01, P<0.05, and P<0.001, respectively). Late addition of Tregs into the effector phase abolished their ability to suppress effector T-cell tissue invasion, resulting in a total loss of their ability to ameliorate GVH tissue damage. CONCLUSION: Preventing effector T-cell tissue invasion is a critical mechanistic event leading to Treg attenuation of GVH tissue damage. This therapeutic effect is associated with a failure of CD8 T cells to increase tissue homing receptors after allo-stimulation, together with a breakdown of interferon-γ-induced chemoattractant expression in the target tissue.


Subject(s)
Chemotaxis, Leukocyte , Graft vs Host Disease/prevention & control , Graft vs Host Reaction/immunology , Skin/immunology , T-Lymphocytes, Regulatory/immunology , Antigens, Ly/metabolism , Cell Adhesion , Cells, Cultured , Chemokine CXCL10/metabolism , Chemokine CXCL11/metabolism , Chemokine CXCL9/metabolism , Coculture Techniques , Flow Cytometry , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Humans , Immunohistochemistry , Interferon-gamma/metabolism , Real-Time Polymerase Chain Reaction , Receptors, CXCR3/metabolism , Skin/pathology , Tissue Culture Techniques
9.
Proc Natl Acad Sci U S A ; 109(12): 4598-603, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22392992

ABSTRACT

The recruitment of T lymphocytes during diseases such as rheumatoid arthritis is regulated by stimulation of the chemokine receptors expressed by these cells. This study was designed to assess the potential of a CXCR3-specific small-molecule agonist to inhibit the migration of activated human T cells toward multiple chemokines. Further experiments defined the molecular mechanism for this anti-inflammatory activity. Analysis in vitro demonstrated agonist induced internalization of both CXCR3 and other chemokine receptors coexpressed by CXCR3(+) T cells. Unlike chemokine receptor-specific antagonists, the CXCR3 agonist inhibited migration of activated T cells toward the chemokine mixture in synovial fluid from patients with active rheumatoid arthritis. A humanized mouse air-pouch model showed that intravenous treatment with the CXCR3 agonist prevented inflammatory migration of activated human T cells toward this synovial fluid. A potential mechanism for this action was defined by demonstration that the CXCR3 agonist induces receptor cross-phosphorylation within CXCR3-CCR5 heterodimers on the surface of activated T cells. This study shows that generalized chemokine receptor desensitization can be induced by specific stimulation of a single chemokine receptor on the surface of activated human T cells. A humanized mouse model was used to demonstrate that this receptor desensitization inhibits the inflammatory response that is normally produced by the chemokines present in synovial fluid from patients with active rheumatoid arthritis.


Subject(s)
Receptors, CXCR3/metabolism , T-Lymphocytes/metabolism , Animals , Arthritis/metabolism , Autoimmunity , Chemokines/metabolism , Female , Flow Cytometry/methods , Humans , Inflammation/pathology , Leukocytes, Mononuclear/cytology , Mice , Mice, Inbred NOD , Phosphorylation , Receptors, CCR5/metabolism , T-Lymphocytes/cytology
10.
Br J Pharmacol ; 166(3): 895-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22035113

ABSTRACT

Chemokines represent a class of cytokines that control the migration of leucocytes. The human chemokine system comprises 44 ligands and 21 receptors that have evolved to control leucocyte migration. Although chemokines are an attractive therapeutic target for anti-inflammatory intervention, clinical trials of small molecule receptor antagonists have failed to demonstrate efficacy. One often cited explanation for this is the apparent redundancy within the chemokine system, wherein several ligands bind and activate each receptor. The work of Scholten et al. and Nedjai et al. reported in this issue of the British Journal of Pharmacology demonstrates that this redundancy does not exist at the molecular level and provides a powerful insight into the complex nature of chemokine receptor activation.


Subject(s)
Chemokine CXCL10/metabolism , Chemokine CXCL11/metabolism , Isoquinolines/pharmacology , Receptors, CXCR3/agonists , Small Molecule Libraries/pharmacology , Animals , Humans
12.
Transplant Rev (Orlando) ; 25(4): 136-44, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21514134

ABSTRACT

More than 45 members of the family of chemotactic cytokines have been described. These chemokines control the migration of leukocytes throughout the whole alloimmune response from initial ischemic damage to acute inflammation and eventual resolution. Several chemokines have been strongly linked to allograft rejection. Recent studies have described powerful endogenous mechanisms that regulate chemokine biology. This review will describe a new class of chemokine receptor that bind ligands with high affinity but lack the capacity for signaling. Atypical receptors represent a new paradigm in chemokine biology and may hold the key to our eventual manipulation of chemokine-driven allograft inflammation.


Subject(s)
Chemokines/immunology , Organ Transplantation , Receptors, Chemokine/immunology , Animals , Anti-Inflammatory Agents/immunology , Graft Rejection/immunology , Humans , Leukocytes/immunology , Signal Transduction , Transplantation, Homologous/immunology
13.
Transplantation ; 89(11): 1411-6, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20404785

ABSTRACT

BACKGROUND: Inflammatory cell recruitment during allograft rejection is driven by a group of inflammatory cytokines termed chemokines. Chemokines are presented on the surface of the vascular endothelium where they ligate specific receptors expressed on the surface of leukocytes. Recently, a group of nonsignaling chemokine receptors have been described. These bind and internalize chemokines but do not drive leukocyte migration. It is believed that these compete with classical signaling receptors to modulate inflammation. METHODS: This study describes the first examination of the human decoy chemokine receptor D6 during rejection; D6 binds at least 12 potent proinflammatory chemokines. The expression of D6 by graft infiltrating leukocytes was examined in cardiac allografts by confocal microscopy on biopsy sections (n=19). Cytokine regulation of D6 was examined in vitro, and a chemokine scavenging assay was performed using the prototypical transplant-associated chemokine CCL5/RANTES. RESULTS: D6 expression was found to be higher in the biopsies taken from more severe cardiac allograft rejection (P<0.01) and was predominantly localized to graft infiltrating CD45(+)CD68(+) leukocytes. In vitro studies demonstrated that the transforming growth factor-beta strongly increased the expression of D6 by monocytes, which significantly enhanced D6-mediated chemokine scavenging (by 85%, P<0.05). CONCLUSIONS: We present the first examination of the biology of D6 during rejection and identify a transplant-associated cytokine that is able to regulate its expression. These data suggest an exciting new mechanism for the antiinflammatory actions of transforming growth factor-beta. Understanding the expression patterns of D6 may provide important insight into the regulation and control of inflammatory cell recruitment during allograft rejection.


Subject(s)
Graft Rejection/immunology , Heart Transplantation/immunology , Receptors, CCR10/genetics , Animals , Biopsy , Cell Line , Cytokines/immunology , Cytokines/physiology , Graft Rejection/pathology , Graft Survival/immunology , Heart Transplantation/pathology , Humans , Leukocytes/physiology , Models, Animal , Monocytes/physiology , Rats , Receptors, CCR10/immunology , Receptors, CCR10/physiology , Receptors, Chemokine/physiology , Transplantation, Homologous/immunology , Chemokine Receptor D6
14.
Hepatology ; 51(6): 1988-97, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20232292

ABSTRACT

UNLABELLED: Acute alcoholic hepatitis is characterized by disproportionate macrophage inflammatory cytokine responses to bacterial lipopolysaccharide. Lack of knowledge of the underlying mechanism has limited progress toward effective therapy. We postulated a novel mechanism by which ethanol increases histone acetylation, increasing proinflammatory gene transcription and cytokine synthesis. Cytokine responses to lipopolysaccharide in a human macrophage cell line cultured in 86 mM ethanol, 1 mM acetate, and normal media were measured by multiplex immunoassay. Changes in histone acetylation were determined by immunofluorescence microscopy and chromatin immunoprecipitation on presentation. The effect of ethanol and acetate on acetyl-coenzyme A (acetyl-coA) synthetases, which convert acetate to acetyl-coA, the substrate for histone acetylation, was determined by quantitative reverse-transcription polymerase chain reaction and immunoblotting. Knockdown of acetyl-coA synthetases by short hairpin RNA (shRNA) was used to determine their role in ethanol's enhancement of the inflammatory cytokine response. Ethanol-exposed macrophages developed enhanced interleukin 6 (IL6), IL8, and tumor necrosis factor alpha responses to lipopolysaccharide with time-dependent increases in histone acetylation that could be prevented by inhibition of ethanol metabolism. Chromatin immunoprecipitation confirmed increased histone acetylation at promoter regions of specific cytokine genes. The effect of ethanol was reproduced by incubation with acetate, the principal hepatic metabolite of ethanol, and both ethanol and acetate reduced histone deacetylase activity and up-regulated acetyl-coA synthetases. Knockdown of the acetyl-coA synthetases abrogated the effect of ethanol on cytokine production. CONCLUSION: Synthesis of metabolically available acetyl-coA from acetate is critical to the increased acetylation of proinflammatory gene histones and consequent enhancement of the inflammatory response in ethanol-exposed macrophages. This mechanism is a potential therapeutic target in acute alcoholic hepatitis.


Subject(s)
Acetates/metabolism , Cytokines/metabolism , Ethanol/metabolism , Gene Expression Regulation , Hepatitis, Alcoholic/metabolism , Macrophages/metabolism , Acetate-CoA Ligase/metabolism , Acetyl Coenzyme A/metabolism , Acetylation , Cell Line , Ethanol/adverse effects , Gene Knockdown Techniques , Hepatitis, Alcoholic/etiology , Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Promoter Regions, Genetic
15.
Mol Immunol ; 47(2-3): 485-92, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19767105

ABSTRACT

Endothelial cells present chemokines to T cells and can also stimulate the T cell antigen receptor by presentation of peptide-MHC antigen complexes. This study was designed to investigate the potential synergy between stimulation of the chemokine receptor CXCR3 and the human T cell receptor complex. Transendothelial T cell migration towards CXCL10 was modified by crosslinking CD3 immediately before addition to the endothelium. When resting endothelium was used, T cells which had been activated by crosslinking CD3 for only 1 min showed a significant reduction (p<0.0001) in migration when compared with untreated T cells. By contrast, endothelial cells which had been activated by stimulation with interferon-gamma and tumour necrosis factor-alpha supported a specific increase in the migration of activated T cells; this was most apparent after CD3 had been activated for 90 min (p<0.0001). The molecular basis for synergy between CXCR3 and the T cell receptor complex was investigated by measurement of fluorescence resonance energy transfer. This showed that CXCL10 induced a close (<10 nm) spatial association between CXCR3 and the CD3epsilon subunit on the cell-surface. These data demonstrate that stimulation of both CXCR3 and the T cell receptor has the potential to enhance specifically both the proliferation and extravasation of specific T cells during episodes of local inflammation.


Subject(s)
Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, CXCR3/immunology , T-Lymphocytes/immunology , Adult , CD3 Complex/immunology , Cell Line , Cell Proliferation/drug effects , Chemotaxis/drug effects , Cross-Linking Reagents/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Fluorescence Resonance Energy Transfer , Humans , Lymphocyte Activation/drug effects , Models, Immunological , Reproducibility of Results , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
16.
FASEB J ; 23(11): 3906-16, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19667120

ABSTRACT

Interaction between chemokines and heparan sulfate (HS) is essential for leukocyte recruitment during inflammation. Previous studies have shown that a non-HS-binding mutant form of the inflammatory chemokine CCL7 can block inflammation produced by wild-type chemokines. This study examined the anti-inflammatory mechanism of a non-HS-binding mutant of the homeostatic chemokine CXCL12. Initial experiments demonstrated that mutant CXCL12 was an effective CXCR4 agonist. However, this mutant chemokine failed to promote transendothelial migration in vitro and inhibited the haptotactic response to wild-type CCL7, CXCL12, and CXCL8, and naturally occurring chemoattractants in synovial fluid from the rheumatoid synovium, including CCL2, CCL7, and CXCL8. Notably, intravenous administration of mutant CXCL12 also inhibited the recruitment of leukocytes to murine air pouches filled with wild-type CXCL12. Following intravenous administration, wild-type CXCL12 was cleared from the circulation rapidly, while the mutant chemokine persisted for >24 h. Chronic exposure to mutant CXCL12 in the circulation reduced leukocyte-surface expression of CXCR4, reduced the chemotactic response of these cells to CXCL12, and inhibited normal chemokine-mediated induction of adhesion between the alpha4beta1 integrin, VLA-4, and VCAM-1. These data demonstrate that systemic administration of non-HS-binding variants of CXCL12 can mediate a powerful anti-inflammatory effect through chemokine receptor desensitization.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chemokine CXCL12/genetics , Receptors, CXCR4/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , CHO Cells , Chemokine CXCL12/administration & dosage , Chemokine CXCL12/pharmacology , Chemotaxis, Leukocyte/immunology , Cricetinae , Cricetulus , Endothelium/metabolism , Female , Heparitin Sulfate/metabolism , Humans , Injections, Intravenous , Integrin alpha4beta1/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Receptors, CXCR4/agonists , Signal Transduction/drug effects
17.
Mol Immunol ; 44(8): 1944-53, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17081610

ABSTRACT

The chemokine receptor CCR2 binds four pro-inflammatory monocyte chemoattractant proteins, designated MCP1/CCL2, MCP2/CCL8, MCP3/CCL7 and MCP4/CCL13. This study demonstrates the important biology of this receptor during the response to the chemokine milieu. Competitive chemotaxis and calcium flux assays were performed utilising mixtures of chemokines to assess a hierarchal arrangement of chemokine prepotency; these demonstrated that the MCP2-CCR2 interaction is able to supersede signals generated by RANTES, another pro-inflammatory chemokine, or the homeostatic chemokine SDF1. These observations were validated using three physiologically relevant monocytic cell lines. Having identified the importance of CCR2, experiments were then performed to examine the signal transduction processes coupled to this receptor. G protein coupling was initially examined; Cholera toxin reduced the chemotactic response to MCP2 (p<0.001), whilst the response to the other MCP chemokines remained normal. The response to MCP2 was uniquely inhibited by elevated concentrations of cAMP and, unlike MCP1, 3 and 4 (p<0.05), MCP2 failed to inhibit adenylate cyclase. Expression of dominant negative H-ras demonstrated that each MCP chemokine required active ras in order to elicit ERK activation and a chemotactic response. Unlike MCP1, MCP2 failed to induce nuclear translocation of activated ERK1 or subsequent induction of c-Myc expression. Akt activation also showed ligand-specific differences, with MCP2 producing a delayed response compared to the other MCP chemokines. Together these data highlight the importance of CCR2 and suggest that it is a powerful tool for fine tuning the immune response.


Subject(s)
Chemotaxis/immunology , Gene Expression Regulation/immunology , Monocyte Chemoattractant Proteins/immunology , Receptors, Chemokine/immunology , Signal Transduction/immunology , Cell Line , Chemotaxis/drug effects , Chemotaxis/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Activation/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Monocyte Chemoattractant Proteins/pharmacology , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , Receptors, CCR2 , Receptors, Chemokine/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
18.
Mol Immunol ; 44(7): 1477-82, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17000001

ABSTRACT

Inflammation plays an important role in a wide range of human diseases. Chemokines are a group of proteins which control the migration and activation of the immune cells involved in all aspects of the inflammatory response. Chemokines bind to specific receptors of the seven-transmembrane spanning type on target leukocytes and also bind to cell-surface glycosaminoglycans (GAG). Leukocytes express a range of chemokine receptors which can cross-desensitise each other, potentially allowing a single chemokine receptor agonist to desensitise all the chemokine receptors on a cell. If an appropriate single receptor agonist is engineered to be non-chemotactic itself, then a treated cell will lose the potential to migrate in response to chemokines towards any developing site of inflammation. A non-GAG-binding but receptor agonistic form of the chemokine CCL7 can inhibit leukocyte recruitment in response to a diverse range of chemokines in vitro and in vivo. We hypothesise that this modified chemokine mediates its effect by inducing homologous and heterologous receptor desensitisation and further propose that other suitable candidates could include agonistic chemokine receptor-specific antibodies or small molecule chemokine receptor agonists. Hence, an appropriate chemokine receptor agonist could be used to inhibit multiple chemokine receptors, thereby producing a powerful and robust anti-inflammatory effect. This review considers the mechanisms leading to chemokine receptor desensitisation and discusses the potential to develop a new class of anti-inflammatory agents based on targeted stimulation of chemokine receptors.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Receptors, Chemokine/agonists , Anti-Inflammatory Agents/pharmacology , Drug Design , Humans , Receptors, Chemokine/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...