Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Commun Biol ; 4(1): 569, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33980979

ABSTRACT

Following the FDA-approval of the hematopoietic stem cell (HSC) mobilizer plerixafor, orally available and potent CXCR4 antagonists were pursued. One such proposition was AMD11070, which was orally active and had superior antagonism in vitro; however, it did not appear as effective for HSC mobilization in vivo. Here we show that while AMD11070 acts as a full antagonist, plerixafor acts biased by stimulating ß-arrestin recruitment while fully antagonizing G protein. Consequently, while AMD11070 prevents the constitutive receptor internalization, plerixafor allows it and thereby decreases receptor expression. These findings are confirmed by the successful transfer of both ligands' binding sites and action to the related CXCR3 receptor. In vivo, plerixafor exhibits superior HSC mobilization associated with a dramatic reversal of the CXCL12 gradient across the bone marrow endothelium, which is not seen for AMD11070. We propose that the biased action of plerixafor is central for its superior therapeutic effect in HSC mobilization.


Subject(s)
Benzylamines/pharmacology , Cyclams/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Receptors, CXCR4/metabolism , Aminoquinolines/metabolism , Aminoquinolines/pharmacology , Animals , Benzimidazoles/metabolism , Benzimidazoles/pharmacology , Benzylamines/metabolism , Butylamines/metabolism , Butylamines/pharmacology , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cyclams/metabolism , Drug Delivery Systems/methods , Female , Granulocyte Colony-Stimulating Factor , HEK293 Cells , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Pharmaceutical Preparations/metabolism , Receptors, CXCR3/drug effects , Receptors, CXCR3/metabolism , Receptors, CXCR4/drug effects , beta-Arrestins/drug effects , beta-Arrestins/metabolism
2.
Peptides ; 93: 44-50, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28526557

ABSTRACT

Frog skin is a source of peptides with various biological properties. Frenatin 2.1S, derived from norepinephrine-stimulated skin secretions of the Orinoco lime tree frog Sphaenorhynchus lacteus, exhibits immunostimulatory effects as demonstrated by the promotion of proinflammatory phenotypes of mononuclear cells in mouse peritoneal cavity and spleen. The aim of this study was to identify the populations of host cells sensitive to the action of frenatin 2.1S in vivo and to study its effects on their functional antitumor capacity. A single injection of frenatin 2.1S (100µg) in BALB/c mice increased the presence of peritoneal CD11c+ dendritic cells and CD3+ T cells 24h after administration and there was a significant increase in the number of IL-17 and CXCR3 expressing inflammatory T cells. Frenatin 2.1S treatment also increased the number of TNF-α expressing F4/80+ proinflammatory M1 macrophages. The most striking finding of the study is the marked increase of the number of peritoneal natural killer (NK) cells following frenatin 2.1S injection. Further, frenatin 2.1S administration led to activation of NK cells as evaluated by increased expression of NKG2D, FasL, CD69 and CD107a. The increased ratio of interferon-γ vs. IL-10 producing NK cells is further indication of the proinflammatory action of frenatin 2.1S. Peptide treatment enhanced the tumoricidal action of peritoneal NK cells on 4T1 mouse mammary carcinoma cells as revealed by the real-time automated monitoring of cell status. Our data demonstrate that frenatin 2.1S promotes activation and cytotoxic capacity of NK cells and should be regarded as a candidate for antitumor immunotherapy.


Subject(s)
Amphibian Proteins/pharmacology , Killer Cells, Natural/drug effects , Peptides/pharmacology , Animals , Anura , Cell Survival/drug effects , Cytokines/analysis , Cytokines/biosynthesis , Cytokines/drug effects , Dendritic Cells/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Male , Mice , Mice, Inbred BALB C , Peritoneal Cavity/cytology , Receptors, CXCR3/drug effects , Statistics, Nonparametric , Tumor Cells, Cultured
3.
Expert Rev Clin Pharmacol ; 9(6): 853-61, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26900630

ABSTRACT

C-X-C chemokine receptor (CXCR)3 and its interferon(IFN)γ-dependent chemokines (CXCL10, CXCL9, CXCL11) are implicated in the immune-pathogenesis of autoimmune thyroiditis (AT), Graves disease (GD) and Graves Ophthalmopathy (GO). In tissue, recruited Th1 lymphocytes produce IFNγ, enhancing the tissue secretion of IFNγ-inducible chemokines, initiating and perpetuating the autoimmune process. Patients with AT (with hypothyroidism), and with GO and GD, particularly in the active phase, have high IFNγ-inducible chemokines. Peroxisome proliferator-activated receptor (PPAR)γ or -α agonists and methimazole exert an immune-modulation on CXCR3 chemokines in AT, GD and GO. Other studies are ongoing to evaluate new molecules acting as antagonists of CXCR3, or blocking CXCL10, in Hashimoto thyroiditis (HT), GD and GO. Recently, novel molecules targeting the various agents involved in the pathogenesis of GO, such as rituximab, have been proposed as an alternative to corticosteroids. However, randomized and controlled studies are needed to generalize these interesting results.


Subject(s)
Graves Disease/drug therapy , Graves Ophthalmopathy/drug therapy , Thyroiditis, Autoimmune/drug therapy , Animals , Chemokines/metabolism , Drug Design , Graves Disease/physiopathology , Graves Ophthalmopathy/physiopathology , Humans , Interferon-gamma/metabolism , Receptors, CXCR3/drug effects , Receptors, CXCR3/metabolism , Thyroiditis, Autoimmune/physiopathology
4.
Clin Sci (Lond) ; 130(7): 463-78, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26888559

ABSTRACT

The CXC chemokines, CXCL4, -9, -10, -11, CXCL4L1, and the CC chemokine CCL21, activate CXC chemokine receptor 3 (CXCR3), a cell-surface G protein-coupled receptor expressed mainly by Th1 cells, cytotoxic T (Tc) cells and NK cells that have a key role in immunity and inflammation. However, CXCR3 is also expressed by vascular smooth muscle and endothelial cells, and appears to be important in controlling physiological vascular function. In the last decade, evidence from pre-clinical and clinical studies has revealed the participation of CXCR3 and its ligands in multiple cardiovascular diseases (CVDs) of different aetiologies including atherosclerosis, hypertension, cardiac hypertrophy and heart failure, as well as in heart transplant rejection and transplant coronary artery disease (CAD). CXCR3 ligands have also proven to be valid biomarkers for the development of heart failure and left ventricular dysfunction, suggesting an underlining pathophysiological relation between levels of these chemokines and the development of adverse cardiac remodelling. The observation that several of the above-mentioned chemokines exert biological actions independent of CXCR3 provides both opportunities and challenges for developing effective drug strategies. In this review, we provide evidence to support our contention that CXCR3 and its ligands actively participate in the development and progression of CVDs, and may additionally have utility as diagnostic and prognostic biomarkers.


Subject(s)
Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , Chemokines, CXC/metabolism , Receptors, CXCR3/metabolism , Signal Transduction , Animals , Biomarkers/blood , Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology , Cardiovascular System/drug effects , Cardiovascular System/pathology , Cardiovascular System/physiopathology , Drug Design , Humans , Ligands , Molecular Targeted Therapy , Predictive Value of Tests , Prognosis , Receptors, CXCR3/drug effects , Signal Transduction/drug effects
5.
Am J Respir Crit Care Med ; 187(1): 65-77, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23144331

ABSTRACT

RATIONALE: Patients who developed acute respiratory distress syndrome (ARDS) after infection with severe respiratory viruses (e.g., severe acute respiratory syndrome-coronavirus, H5N1 avian influenza virus), exhibited unusually high levels of CXCL10, which belongs to the non-ELR (glutamic-leucine-arginine) CXC chemokine superfamily. CXCL10 may not be a bystander to the severe virus infection but may directly contribute to the pathogenesis of neutrophil-mediated, excessive pulmonary inflammation. OBJECTIVES: We investigated the contribution of CXCL10 and its receptor CXCR3 axis to the pathogenesis of ARDS with nonviral and viral origins. METHODS: We induced nonviral ARDS by acid aspiration and viral ARDS by intratracheal influenza virus infection in wild-type mice and mice deficient in CXCL10, CXCR3, IFNAR1 (IFN-α/ß receptor 1), or TIR domain-containing adaptor inducing IFN-ß (TRIF). MEASUREMENTS AND MAIN RESULTS: We found that the mice lacking CXCL10 or CXCR3 demonstrated improved severity and survival of nonviral and viral ARDS, whereas mice that lack IFNAR1 did not control the severity of ARDS in vivo. The increased levels of CXCL10 in lungs with ARDS originate to a large extent from infiltrated pulmonary neutrophils, which express a unique CXCR3 receptor via TRIF. CXCL10-CXCR3 acts in an autocrine fashion on the oxidative burst and chemotaxis in the inflamed neutrophils, leading to fulminant pulmonary inflammation. CONCLUSIONS: CXCL10-CXCR3 signaling appears to be a critical factor for the exacerbation of the pathology of ARDS. Thus, the CXCL10-CXCR3 axis could represent a prime therapeutic target in the treatment of the acute phase of ARDS of nonviral and viral origins.


Subject(s)
Chemokine CXCL10/physiology , Lung Injury/physiopathology , Neutrophils/physiology , Orthomyxoviridae Infections/physiopathology , Receptors, CXCR3/physiology , Respiratory Distress Syndrome/physiopathology , Aged , Aged, 80 and over , Animals , Chemokine CXCL10/drug effects , Disease Models, Animal , Disease Progression , Humans , Influenza A Virus, H5N1 Subtype , Lung Injury/immunology , Lung Injury/virology , Male , Mice , Mice, Inbred Strains , Middle Aged , Neutrophils/immunology , Orthomyxoviridae Infections/immunology , Protein Array Analysis , Rats , Rats, Sprague-Dawley , Receptors, CXCR3/drug effects , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/virology
6.
Transplantation ; 93(1): 24-31, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22124337

ABSTRACT

BACKGROUND: The chemokine receptors CCR5 and CXCR3 are expressed by T cells and macrophages. We examined effects of a CCR5/CXCR3 antagonist (TAK), with a particular focus on the role of macrophages, in a rat kidney transplant model. METHODS: Dark Agouti rat kidneys were transplanted into Lewis rats. The recipients were treated daily with a 10 mg/kg TAK on posttransplant days 0 to 14 and/or 2 mg/kg of cyclosporine A (CsA) on days 0 to 5. Graft survival, histological changes, and the expression of chemokines and chemokine receptors on T cells and macrophages were studied. RESULTS: Treatment with TAK alone suppressed CD4+T cell infiltration and slightly prolonged graft survival. The expressions of both CCR5 and CXCR3, and activated macrophage-associated cytokines and chemokines, were significantly increased on macrophages that had been separated from rejecting kidneys, compared with those from spleens. However, these upregulations were decreased in macrophages from kidneys that had been treated with TAK. Immunohistochemistry also showed that macrophages infiltrating tubules of rejecting kidney expressed both receptors. In the CsA alone group, macrophages were the dominant infiltrating cells, and all allografts were rejected within 10 days. A combined therapy involving CsA and TAK resulted in decreased macrophage infiltration, and graft survival was substantially prolonged. The levels of activated macrophage-associated cytokines and chemokines were also decreased. CONCLUSION: The dual blocking of CCR5/CXCR3 can be useful in decreasing rejection, with or without CsA. This mechanism acts, not only to block T-cell recruitment to a kidney graft but to suppress the infiltration of macrophages as well.


Subject(s)
CCR5 Receptor Antagonists , Cell Movement/physiology , Graft Rejection/pathology , Graft Rejection/physiopathology , Kidney Transplantation/pathology , Macrophages/physiology , Receptors, CXCR3/antagonists & inhibitors , Amides/pharmacology , Animals , Chemokines/metabolism , Cyclosporine/therapeutic use , Cytokines/metabolism , Graft Rejection/prevention & control , Graft Survival/drug effects , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/immunology , Male , Models, Animal , Quaternary Ammonium Compounds/pharmacology , Rats , Rats, Inbred Lew , Rats, Inbred Strains , Receptors, CCR5/drug effects , Receptors, CCR5/physiology , Receptors, CXCR3/drug effects , Receptors, CXCR3/physiology , Transplantation, Homologous
7.
Eur J Immunol ; 39(1): 113-25, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19130557

ABSTRACT

We examined the hypothesis that stromal fibroblasts modulate the ability of endothelial cells (EC) to recruit lymphocytes in a site-specific manner. PBL were perfused over HUVEC that had been cultured with fibroblasts isolated from the inflamed synovium or the skin of patients with rheumatoid arthritis or osteoarthritis, or from normal synovium, with or without exposure to the inflammatory cytokines TNF-alpha+IFN-gamma. Fibroblasts from inflamed synovium, but no others, caused unstimulated HUVEC to bind flowing lymphocytes. This adhesion was supported by alpha(4)beta(1)-VCAM-1 interaction and stabilised by activation of PBL through CXCR4-CXCL12. Antibody neutralisation of IL-6 during co-culture effectively abolished the ability of EC to bind lymphocytes. Cytokine-stimulated EC supported high levels of lymphocyte adhesion, through the presentation of VCAM-1, E-selectin and chemokine(s) acting through CXCR3. Interestingly, co-culture with dermal fibroblasts caused a marked reduction in cytokine-induced adhesion, while synovial fibroblasts had variable effects depending on their source. In the dermal co-cultures, neutralisation of IL-6 or TGF-beta caused partial recovery of cytokine-induced lymphocyte adhesion; this was complete when both were neutralised. Exogenous IL-6 was also found to inhibit response to TNF-alpha+IFN-gamma. Normal stromal fibroblasts appear to regulate the cytokine-sensitivity of vascular endothelium, while fibroblasts associated with chronic inflammation bypass this and develop a directly inflammatory phenotype. Actions of IL-6 might be pro-inflammatory or anti-inflammatory, depending on the local milieu.


Subject(s)
Cell Movement/immunology , Endothelial Cells/immunology , Fibroblasts/immunology , Inflammation/immunology , Lymphocytes/immunology , Antiviral Agents/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cells, Cultured , Chemokine CXCL12/immunology , Chemokine CXCL12/metabolism , Coculture Techniques , E-Selectin/immunology , E-Selectin/metabolism , Endothelial Cells/drug effects , Fibroblasts/drug effects , Humans , Interferon-gamma/pharmacology , Interleukin-6/antagonists & inhibitors , Interleukin-6/immunology , Interleukin-6/metabolism , Interleukin-6/pharmacology , Lymphocytes/drug effects , Oligonucleotide Array Sequence Analysis , Receptors, CXCR3/drug effects , Receptors, CXCR3/immunology , Receptors, CXCR3/metabolism , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Synovial Membrane/immunology , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism
8.
Transplant Proc ; 40(5): 1300-2, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18589091

ABSTRACT

OBJECTIVE: To explore the expression of the CXC chemokine receptor 3 (CXCR3) and its ligands (IP-10, Mig) in ischemiareperfusion (I/R) injury of rat livers. METHODS: Thirty-two Wistar rats were randomly divided into four groups with eight rats in each group: sham operation (SO) and 6-, 12-, or 24-hour I/R groups. The levels of tumor necrosis factor-alpha (TNF-alpha) in liver tissues were measured by enzyme-linked immunosorbent assay. The expressions of CXCR3 and its ligands (IP-10, Mig) were detected by semiquantitative reverse-transcriptase polymerase chain reaction. The serum levels of alanine transferase and aspartate transferase were also measured. RESULTS: Low expressions of CXCR3, IP-10, and Mig mRNA were determined in the SO group. The expressions of CXCR3 and IP-10 mRNA in the ischemic tissue of the I/R group were significantly greater than those in the SO group (P < .01). The expressions of CXCR3 and IP-10 mRNA of the ischemic tissue in the 6-hour I/R group were higher than those in the 12-hour I/R group (P < .01). There was no difference in the levels of Mig mRNA between the I/R and SO groups. Compared with the SO group, the level of TNF-alpha was significantly increased in the I/R group, reaching its peak at reperfusion 12 hour. CONCLUSION: The mRNA expressions of CXCR3 and its ligand IP-10 were up-regulated in liver I/R tissue in the early time, which suggested that they play an important role in liver injury induced by I/R.


Subject(s)
Liver Circulation , Receptors, CXCR3/genetics , Reperfusion Injury/physiopathology , Animals , Gene Expression Regulation/physiology , Rats , Rats, Wistar , Receptors, CXCR3/drug effects , Reverse Transcriptase Polymerase Chain Reaction
10.
Eur Respir J ; 31(4): 783-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18094012

ABSTRACT

Asthma is associated with increased numbers of T-cells in the lung. CC chemokine receptor (CCR)5 and CXC chemokine receptor (CXCR)3 have been reported to play important roles in the lung T-cell homing pathway, and may be potential targets for asthma therapy. The aim of the present study was to investigate the role of CCR5 and CXCR3 in allergen-induced acute asthma and to determine whether a novel small-molecule compound, TAK-779, targeting CCR5 and CXCR3 can attenuate allergic airway responses. Mice were sensitised with ovalbumin (OVA). mRNA expression of chemokine receptors in the lung were measured after the challenge with either aerosolised phosphate-buffered saline or OVA. OVA-sensitised mice were also treated with TAK-779. Respiratory function was measured, bronchoalveolar lavage was performed, and blood and lung samples were obtained. OVA challenge increased CCR3, CCR5 and CXCR3 expression in the lung. Treatment with TAK-779 significantly attenuated altered respiratory function and pulmonary allergic inflammation. The beneficial effect was associated with reduced expression of CCR5 and CXCR3 in the lung. These data demonstrate that blockade of CC chemokine receptor 5 and CXC chemokine receptor 3 using TAK-779, a synthetic nonpeptide compound, can prevent the development of asthma features in a mouse model. Thus, CC chemokine receptor 5 and CXC chemokine receptor 3 may be potential targets for asthma therapy.


Subject(s)
Amides/pharmacology , Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Asthma/immunology , Quaternary Ammonium Compounds/pharmacology , Receptors, CCR5 , Receptors, CXCR3 , Animals , Disease Models, Animal , Female , Immunization , Mice , Mice, Inbred BALB C , Ovalbumin , Receptors, CCR/antagonists & inhibitors , Receptors, CCR3/immunology , Receptors, CCR5/drug effects , Receptors, CCR5/immunology , Receptors, CXCR3/drug effects , Receptors, CXCR3/immunology
11.
J Leukoc Biol ; 83(1): 212-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17947392

ABSTRACT

Hyperforin (Hyp) is an active compound contained in the extract of Hypericum perforatum, well known for its antidepressant activity. However, Hyp has been found to possess several other biological properties, including inhibitory effects on tumor invasion, angiogenesis, and inflammation. In this paper, we show that treatment with Hyp inhibited IFN-gamma production, with down-regulation of T-box (T-bet; marker of Th1 gene expression) and up-regulation of GATA-3 (marker gene of Th2) on IL-2/PHA-activated T cells. In parallel, we showed a strong down-regulation of the chemokine receptor CXCR3 expression on activated T cells. The latter effect and the down-modulation of matrix metalloproteinase 9 expression may eventually lead to the inhibition of migratory capability and matrix traversal toward the chemoattractant CXCL10 by activated lymphocytes that we observed in vitro. The effect of Hyp was thus evaluated on an animal model of experimental allergic encephalomyelitis (EAE), a classic, Th1-mediated autoimmune disease of the CNS, and we observed that Hyp attenuates the severity of the disease symptoms significantly. Together, these properties qualify Hyp as a putative, therapeutic molecule for the treatment of autoimmune inflammatory disease sustained by Th1 cells, including EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Phloroglucinol/analogs & derivatives , T-Lymphocytes/drug effects , Terpenes/pharmacology , Th1 Cells/drug effects , Animals , Bridged Bicyclo Compounds/pharmacology , Bridged Bicyclo Compounds/therapeutic use , Cell Survival/drug effects , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , GATA3 Transcription Factor/drug effects , GATA3 Transcription Factor/metabolism , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-2/pharmacology , Phloroglucinol/pharmacology , Phloroglucinol/therapeutic use , Phytohemagglutinins/pharmacology , Rats , Rats, Inbred Lew , Receptors, CXCR3/biosynthesis , Receptors, CXCR3/drug effects , T-Box Domain Proteins/drug effects , T-Box Domain Proteins/metabolism , T-Lymphocytes/immunology , Terpenes/therapeutic use , Th1 Cells/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
12.
Arterioscler Thromb Vasc Biol ; 28(2): 251-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18048768

ABSTRACT

OBJECTIVE: The chemokine receptor CXCR3 is implicated in migration of leukocytes to sites of inflammation. Antagonizing CXCR3 may be a strategy to inhibit inflammation-induced leukocyte migration and subsequently reduce atherosclerosis. We used the CXCR3 specific antagonist NBI-74330 to block CXCR3-mediated signaling in peritonitis and diet-induced atherosclerosis. METHODS AND RESULTS: Antagonizing CXCR3 with NBI-74330 resulted in a significant reduction in CD4+ T cell and macrophage migration to the peritoneal cavity, which was as shown in ex vivo migration studies totally CXCR3 dependent. Atherosclerotic lesion formation in the aortic valve leaflet area and the entire aorta was significantly inhibited in NBI-74330 treated mice. Lymph nodes draining from the aortic arch were significantly smaller in treated mice and were enriched in regulatory T cells and contained fewer activated T cells, whereas the markers for regulatory T cells within the lesion were enhanced after NBI-74330 treatment. CONCLUSIONS: This study shows for the first time that treatment with a CXCR3 antagonist results in attenuating atherosclerotic lesion formation by blocking direct migration of CXCR3+ effector cells from the circulation into the atherosclerotic plaque and by beneficially modulating the inflammatory response in the lesion and the lymph nodes draining from the atherosclerotic lesion.


Subject(s)
Acetamides/pharmacology , Atherosclerosis/drug therapy , Cell Movement/drug effects , Pyrimidines/pharmacology , Receptors, CXCR3/antagonists & inhibitors , Receptors, LDL/deficiency , Animals , Atherosclerosis/physiopathology , Diet, Atherogenic , Disease Models, Animal , Female , Lymph Nodes/drug effects , Lymph Nodes/physiopathology , Macrophages/drug effects , Mice , Mice, Knockout , Receptors, CXCR3/drug effects , Receptors, CXCR3/physiology , T-Lymphocytes, Regulatory/drug effects
13.
Mini Rev Med Chem ; 7(11): 1089-96, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18045212

ABSTRACT

CXCR3 and CCR5 are chemokine receptor that are predominantly expressed on the surface of Th1 polarized T cells. In a variety of human and experimental autoimmune diseases the enhanced expression of CXCR3 and CCR5 binding chemokine ligands is followed by the recruitment of CXCR3- and CCR5-positive T cells, indicating an important role for these chemokine receptors in T cell-mediated tissue damage. In this review, we summarize a number of in vivo studies available on the neutralization of CXCR3 and CCR5 in inflammatory disease, and specifically focus on the potential therapeutic effects of CXCR3 and CCR5 blockade in human autoimmune disease and organ transplantation.


Subject(s)
Autoimmune Diseases/therapy , Drug Delivery Systems , Receptors, CCR5/metabolism , Receptors, CXCR3/metabolism , Th1 Cells/immunology , Animals , Autoimmune Diseases/immunology , Humans , Ligands , Receptors, CCR5/drug effects , Receptors, CXCR3/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...