Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Food Chem Toxicol ; 176: 113783, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37059382

RESUMEN

Polycyclic aromatic hydrocarbons (PAHs) exposure in food is closely associated with the occurrence and development of breast cancer, which may attribute to altered immunotoxicity and immune regulation. Currently, cancer immunotherapy aims to promote tumor-specific T cell responses, especially CD4+T helper cells (Th) for anti-tumor immunity. The histone deacetylase inhibitors (HDACis) are found to exert an anti-tumor effect by reshaping the tumor immune microenvironment, but the immune regulatory mechanism of HDACis in PAHs-induced breast tumor remains elusive. Here, using established breast cancer models induced by 7,12-dimethylbenz[a]anthracene (DMBA), a potent carcinogenic agent of PAH, the novel HDACi, 2-hexyl-4-pentylene acid (HPTA) exhibited anti-tumor effect by activating T lymphocytes immune function. HPTA recruited CXCR3+CD4+T cells into chemokines CXCL9/10-enriched tumor sites, and the increased secretion of CXCL9/10 was regulated by the NF-κB-mediated pathway. Furthermore, HPTA promoted Th1 differentiation and assisted cytotoxic CD8+T cells in the elimination of breast cancer cells. These findings support the proposition of HPTA as a potential therapeutic in the treatment of PAHs-induced carcinogenicity.


Asunto(s)
Neoplasias de la Mama , Hidrocarburos Policíclicos Aromáticos , Humanos , Femenino , Linfocitos T , Hidrocarburos Policíclicos Aromáticos/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Carcinógenos/metabolismo , Microambiente Tumoral , Quimiocina CXCL9/metabolismo , Quimiocina CXCL9/farmacología , Receptores CXCR3/metabolismo
2.
Aging (Albany NY) ; 12(1): 502-517, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31913856

RESUMEN

Chemokines play essential roles in the progression of various human cancers; however, the expression and role of CXC chemokines in pancreatic adenocarcinoma (PAAD) have not yet been identified. The aim of this study is to identify the expression patterns, clinical significance and mechanisms of CXC chemokines in regulating tumour microenvironment of PAAD. Three CXC chemokines, including CXCL5, CXCL9, and CXCL10, were significantly overexpressed in PAAD tissues, which were correlated with the poor survival of the patients. CXCL9/10 was associated with change of immune cell pattern in the tumour microenvironment, and supplementation of CXCL9 in the orthotopic murine PAAD model promoted tumour progression. In particular, CXCL9 reduced the CD8+ cytotoxic T lymphocytes in the tumour microenvironment of PAAD, which could be attributed to the reduced CD8+ T cell proliferation, activation, and secretion of anti-tumour cytokines. In vitro treatment of CXCL9 directly led to the suppression of the proliferation, activation, and secretion of anti-tumour cytokines of isolated CD8+ T cells. Inhibition of STAT3 recovered the CXCL9-inhibited proliferation, activation, and secretion of anti-tumour cytokines of CD8+ T cells. Our study indicates CXCL9 as a potential target of immunotherapy in PAAD treatment by regulating the CD8+ T lymphocytes in the tumour microenvironment.


Asunto(s)
Adenocarcinoma/inmunología , Adenocarcinoma/metabolismo , Quimiocina CXCL9/metabolismo , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Factor de Transcripción STAT3/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Quimiocina CXCL9/genética , Quimiocina CXCL9/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Expresión Génica , Humanos , Inmunomodulación , Inmunofenotipificación , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Ratones , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Pronóstico , Proteínas Recombinantes/farmacología , Transducción de Señal , Linfocitos T Citotóxicos/efectos de los fármacos , Microambiente Tumoral/inmunología
3.
J Leukoc Biol ; 104(2): 413-422, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29733455

RESUMEN

This study investigates if treatment with a peptide corresponding to the 30 C-terminal amino acids of CXCL9, CXCL9(74-103), ameliorates joint inflammation in a murine model of antigen-induced arthritis (AIA). AIA was induced in male C57BL/6J mice. Intravenous injection of CXCL9(74-103), simultaneously performed with a tibiofemoral challenge with methylated BSA (mBSA) as antigen in mice immunized with mBSA, diminished the accumulation of leukocytes, in particular neutrophils, in the synovial cavity. The levels of the chemokines CXCL1, CXCL2, and CXCL6 and of the cytokine IL-6 were decreased in inflamed periarticular tissue of mice treated with the CXCL9-derived peptide compared to non-treated AIA mice. In addition, CXCL9(74-103) treatment substantially reduced joint and cartilage damage. CXCL9(74-103) competes with CXCL6 and CCL3 for binding to the glycosaminoglycans heparan sulfate and chondroitin sulfate in vitro. In vivo, CXCL9(74-103) quickly binds to blood vessels in joints as observed by confocal microscopy. Next, we evaluated if later treatment with CXCL9(74-103) had a beneficial impact on joint inflammation. CXCL9(74-103) injection 6 h after mBSA challenge still reduced neutrophil accumulation in the joint, although it did not reduce chemokine and IL-6 concentrations. However, a delay of treatment until 12 h after challenge had no effect on cell recruitment and chemokine and IL-6 levels. Taken together, we demonstrated that treatment with a peptide, which interferes with the interaction between chemokines and glycosaminoglycans, from the beginning of the disease controlled the massive accumulation of neutrophils in the joint of AIA mice, greatly impacting on joint inflammation and tissue damage.


Asunto(s)
Artritis Experimental/patología , Artritis Reumatoide/patología , Quimiocina CXCL9/farmacología , Infiltración Neutrófila/efectos de los fármacos , Animales , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Péptidos/farmacología
4.
J Neuroinflammation ; 14(1): 103, 2017 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-28486971

RESUMEN

BACKGROUND: Hypomethylation of the cathepsin Z locus has been proposed as an epigenetic risk factor for multiple sclerosis (MS). Cathepsin Z is a unique lysosomal cysteine cathepsin expressed primarily by antigen presenting cells. While cathepsin Z expression has been associated with neuroinflammatory disorders, a role for cathepsin Z in mediating neuroinflammation has not been previously established. METHODS: Experimental autoimmune encephalomyelitis (EAE) was induced in both wildtype mice and mice deficient in cathepsin Z. The effects of cathepsin Z-deficiency on the processing and presentation of the autoantigen myelin oligodendrocyte glycoprotein, and on the production of IL-1ß and IL-18 were determined in vitro from cells derived from wildtype and cathepsin Z-deficient mice. The effects of cathepsin Z-deficiency on CD4+ T cell activation, migration, and infiltration to the CNS were determined in vivo. Statistical analyses of parametric data were performed by one-way ANOVA followed by Tukey post-hoc tests, or by an unpaired Student's t test. EAE clinical scoring was analyzed using the Mann-Whitney U test. RESULTS: We showed that mice deficient in cathepsin Z have reduced neuroinflammation and dramatically lowered circulating levels of IL-1ß during EAE. Deficiency in cathepsin Z did not impact either the processing or the presentation of MOG, or MOG- specific CD4+ T cell activation and trafficking. Consistently, we found that cathepsin Z-deficiency reduced the efficiency of antigen presenting cells to secrete IL-1ß, which in turn reduced the ability of mice to generate Th17 responses-critical steps in the pathogenesis of EAE and MS. CONCLUSION: Together, these data support a novel role for cathepsin Z in the propagation of IL-1ß-driven neuroinflammation.


Asunto(s)
Catepsina Z/metabolismo , Encefalomielitis Autoinmune Experimental/complicaciones , Epilepsia/etiología , Animales , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/patología , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/patología , Catepsina Z/genética , Quimiocina CXCL9/farmacología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/cirugía , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Leucocitos/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Glicoproteína Mielina-Oligodendrócito/metabolismo , Glicoproteína Mielina-Oligodendrócito/toxicidad , Fragmentos de Péptidos/toxicidad , Fagosomas/metabolismo , Médula Espinal/patología
5.
Oncotarget ; 7(12): 14405-14, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-26883105

RESUMEN

Metastasis of liver cancer is closely linked to tumor microenvironment, in which chemokines and their receptors act in an important role. The CXCR3, the receptor of chemokine CXCL9, belongs to a superfamily of rhodopsin-like seven transmembrane GPCRs and CXCR subfamily. In HCC tissues, CXCR3 was frequently upregulated and correlated with tumor size, tumor differentiation, portal invasion and metastasis. In the study, CXCR3-A isoform that was bound by CXCL9 was found to cause significant change of ERK1/2 phosphorylation level in the MAPK signaling pathway, consequently upregulating the MMP2 and MMP9 expression and promoting invasion and metastasis of CD133+ liver cancer cells. Also, CXCR3-A suppressed the adhesion ability of CD133+ liver cancer cells that stimulated by CXCL9 for 24h. These findings suggest that CXCR3 and its ligand CXCL9 could promote the metastasis of liver cancer cells and might be a potential target for the intervention of liver cancer metastasis.


Asunto(s)
Antígeno AC133/metabolismo , Empalme Alternativo , Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/secundario , Quimiocina CXCL9/farmacología , Neoplasias Hepáticas/patología , Receptores CXCR3/genética , Animales , Apoptosis , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Movimiento Celular , Proliferación Celular , Femenino , Estudios de Seguimiento , Humanos , Interferón gamma/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Persona de Mediana Edad , Invasividad Neoplásica , Estadificación de Neoplasias , Pronóstico , Células Tumorales Cultivadas , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Haematologica ; 101(3): e99-102, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26589908
7.
Biochem Pharmacol ; 100: 73-85, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26551597

RESUMEN

Chemokines attract leukocytes to sites of infection in a G protein-coupled receptor (GPCR) and glycosaminoglycan (GAG) dependent manner. Therefore, chemokines are crucial molecules for proper functioning of our antimicrobial defense mechanisms. In addition, some chemokines have GPCR-independent defensin-like antimicrobial activities against bacteria and fungi. Recently, high affinity for GAGs has been reported for the positively charged COOH-terminal region of the chemokine CXCL9. In addition to CXCL9, also CXCL12γ has such a positively charged COOH-terminal region with about 50% positively charged amino acids. In this report, we compared the affinity of COOH-terminal peptides of CXCL9 and CXCL12γ for GAGs and KD values in the low nM range were detected. Several enveloped viruses such as herpesviruses, hepatitis viruses, human immunodeficiency virus (HIV), dengue virus (DENV), etc. are known to bind to GAGs such as the negatively charged heparan sulfate (HS). In this way GAGs are important for the initial contacts between viruses and host cells and for the infection of the cell. Thus, inhibiting the virus-cell interactions, by blocking GAG-binding sites on the host cell, might be a way to target multiple virus families and resistant strains. This article reports that the COOH-terminal peptides of CXCL9 and CXCL12γ have antiviral activity against DENV serotype 2, clinical and laboratory strains of herpes simplex virus (HSV)-1 and respiratory syncytial virus (RSV). Moreover, we show that CXCL9(74-103) competes with DENV envelope protein domain III for binding to heparin. These short chemokine-derived peptides may be lead molecules for the development of novel antiviral agents.


Asunto(s)
Antivirales/metabolismo , Quimiocinas/metabolismo , Virus del Dengue/metabolismo , Glicosaminoglicanos/metabolismo , Herpesvirus Humano 1/metabolismo , Virus Sincitiales Respiratorios/metabolismo , Secuencia de Aminoácidos , Animales , Antivirales/farmacología , Células CHO , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/farmacología , Quimiocina CXCL9/genética , Quimiocina CXCL9/metabolismo , Quimiocina CXCL9/farmacología , Quimiocinas/genética , Quimiocinas/farmacología , Cricetinae , Cricetulus , Virus del Dengue/efectos de los fármacos , Glicosaminoglicanos/farmacología , Células HeLa , Herpesvirus Humano 1/efectos de los fármacos , Humanos , Datos de Secuencia Molecular , Unión Proteica , Virus Sincitiales Respiratorios/efectos de los fármacos
8.
J Immunol ; 195(6): 2788-96, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26268659

RESUMEN

Epithelial to mesenchymal cell transition (EMT), whereby fully differentiated epithelial cells transition to a mesenchymal phenotype, has been implicated in the pathogenesis of idiopathic pulmonary fibrosis (IPF). CXCR3 and its ligands are recognized to play a protective role in pulmonary fibrosis. In this study, we investigated the presence and extent of EMT and CXCR3 expression in human IPF surgical lung biopsies and assessed whether CXCR3 and its ligand CXCL9 modulate EMT in alveolar epithelial cells. Coexpression of the epithelial marker thyroid transcription factor-1 and the mesenchymal marker α-smooth muscle actin and CXCR3 expression was examined by immunohistochemical staining of IPF surgical lung biopsies. Epithelial and mesenchymal marker expression was examined by quantitative real-time PCR, Western blotting, and immunofluorescence in human alveolar epithelial (A549) cells treated with TGF-ß1 and CXCL9, with Smad2, Smad3, and Smad7 expression and cellular localization examined by Western blotting. We found that significantly more cells were undergoing EMT in fibrotic versus normal areas of lung in IPF surgical lung biopsy samples. CXCR3 was expressed by type II pneumocytes and fibroblasts in fibrotic areas in close proximity to cells undergoing EMT. In vitro, CXCL9 abrogated TGF-ß1-induced EMT. A decrease in TGF-ß1-induced phosphorylation of Smad2 and Smad3 occurred with CXCL9 treatment. This was associated with increased shuttling of Smad7 from the nucleus to the cytoplasm where it inhibits Smad phosphorylation. This suggests a role for EMT in the pathogenesis of IPF and provides a novel mechanism for the inhibitory effects of CXCL9 on TGF-ß1-induced EMT.


Asunto(s)
Quimiocina CXCL9/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Fibrosis Pulmonar Idiopática/patología , Mucosa Respiratoria/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Actinas/biosíntesis , Biomarcadores/metabolismo , Línea Celular , Quimiocina CXCL9/farmacología , Células Epiteliales/metabolismo , Humanos , Proteínas Nucleares/biosíntesis , Fosforilación , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Receptores CXCR3/biosíntesis , Receptores CXCR3/metabolismo , Mucosa Respiratoria/citología , Proteína Smad2/biosíntesis , Proteína smad3/biosíntesis , Proteína smad7/biosíntesis , Factor Nuclear Tiroideo 1 , Factores de Transcripción/biosíntesis , Factor de Crecimiento Transformador beta1/farmacología
9.
PLoS One ; 8(3): e58129, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23533582

RESUMEN

Chemokines and chemokine receptor-mediated effects are important mediators of the immunological response and cure in human leishmaniasis. However, in addition to their signalling properties for leukocytes, many chemokines have also been shown to act directly as antimicrobial peptides on bacteria and fungi. We screened ten human chemokines (CXCL2, CXCL6, CXCL8, CXCL9, CXCL10, CCL2, CCL3, CCL20, CCL27, CCL28) for antimicrobial effects on the promastigote form of the protozoan parasite Leishmania mexicana, and observed direct parasiticidal effects of several, CCL28 being the most potent. Damage to the plasma membrane integrity could be visualised by entrance of propidium iodide, as measured with flow cytometry, and by scanning electron microscopy, which showed morphological changes and aggregation of cells. The findings were in concordance with parasiticidal activity, measured by decreased mitochondrial activity in an MTT-assay. This is the first report of direct antimicrobial activity by chemokines on parasites. This component of immunity against Leishmania parasites identified here warrants further investigation that might lead to new insight in the mechanisms of human infection and/or new therapeutic approaches.


Asunto(s)
Antiinfecciosos/farmacología , Antiparasitarios/farmacología , Quimiocinas/farmacología , Leishmania mexicana/efectos de los fármacos , Péptidos/farmacología , Quimiocina CCL2/farmacología , Quimiocina CCL20/farmacología , Quimiocina CCL27/farmacología , Quimiocina CCL3/farmacología , Quimiocina CXCL10/farmacología , Quimiocina CXCL2/farmacología , Quimiocina CXCL6/farmacología , Quimiocina CXCL9/farmacología , Humanos , Interleucina-8/farmacología
10.
J Immunol ; 190(7): 3696-705, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23447686

RESUMEN

The chemokine receptors CXCR3 and CXCR4 are primarily involved in memory Th1 cell-driven autoimmune diseases. Although recent studies in chronic inflammatory disease showed therapeutic success using combined blockade, details of CXCR3 and CXCR4 synergism are not understood. In this investigation, we intended to unravel the interaction of these chemokine receptors in static and dynamic cell-migration assays at both the cellular and molecular levels. Effects of combined stimulation by murine CXCL9 and CXCL12, ligands of CXCR3 and CXCR4, respectively, were analyzed using a murine central memory Th1 cell clone. Costimulation with CXCL9 desensitized the chemotaxis of Th1 cells toward CXCL12 by up to 54%. This effect was found in murine EL-4 cells, as well as in primary human T cells. Furthermore, under dynamic flow conditions CXCL12-induced crawling and endothelial transmigration of Th1 cells was desensitized by CXCL9. Subsequent experiments uncovered several molecular mechanisms underlying the heterologous cross-regulation of CXCR4 signaling by the CXCR3 ligand. CXCR4 surface expression was reduced, whereas CXCL12-induced Akt phosphorylation and intracellular Ca(2+) signals were modulated. Moreover, blockade of Rac by NSC23766 revealed differential effects on CXCL12 and CXCL9 chemotaxis and abolished the desensitizing effect of CXCL9. The desensitization of CXCR4 via CXCR3 in memory Th1 cells suggests that their in vivo homeostasis, widely regulated by CXCL12, seemed to be significantly altered by CXCR3 ligands. Our data provide a more detailed understanding for the continuing extravasation and recruitment of Th1 lymphocytes into sites of persistent inflammation.


Asunto(s)
Quimiocina CXCL12/metabolismo , Quimiocina CXCL9/metabolismo , Memoria Inmunológica , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Calcio/metabolismo , Quimiocina CXCL12/farmacología , Quimiocina CXCL9/farmacología , Quimiotaxis/efectos de los fármacos , Quimiotaxis/inmunología , Humanos , Activación de Linfocitos/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores CXCR3/metabolismo , Receptores CXCR4/metabolismo , Linfocitos T/efectos de los fármacos , Células TH1/efectos de los fármacos , Células TH1/inmunología , Migración Transendotelial y Transepitelial/efectos de los fármacos , Migración Transendotelial y Transepitelial/inmunología , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Proteínas de Unión al GTP rac/metabolismo
11.
Biochem Biophys Res Commun ; 419(2): 412-8, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22349504

RESUMEN

The chemokine receptor CXCR3 is a G-protein-coupled receptor that signals through the Gα(i) class of heterotrimeric G-proteins. CXCR3 is highly expressed on activated T cells and has been proposed to be a therapeutic target in autoimmune disease. CXCR3 is activated by the chemokines CXCL9, CXCL10 and CXCL11. CXCR3 signaling properties in response to CXCL10, CXCL11 and the synthetic agonist VUF10661 have previously been evaluated using conventional endpoint assays. In the present study, label-free impedance measurements were used to characterize holistic responses of CXCR3-expressing cells to stimulation with chemokines and VUF10661 in real time and to compare these responses with both G-protein and non-G-protein (ß-arrestin2) mediated responses. Differences in response kinetics were apparent between the chemokines and VUF10661. Moreover, CXCR3-independent effects could be distinguished from CXCR3-specific responses with the use of the selective CXCR3 antagonist NBI-74330 and the Gα(i) inhibitor pertussis toxin. By comparing the various responses, we observed that CXCL9 is a biased CXCR3 agonist, stimulating solely G-protein-dependent pathways. Moreover, CXCR3-mediated changes in cellular impedance correlated with G-protein signaling, but not ß-arrestin2 recruitment.


Asunto(s)
Impedancia Eléctrica , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/agonistas , Isoquinolinas/farmacología , Receptores CXCR3/agonistas , Acetamidas/farmacología , Quimiocina CXCL10/farmacología , Quimiocina CXCL11/farmacología , Quimiocina CXCL9/farmacología , Células HEK293 , Humanos , Ligandos , Redes y Vías Metabólicas , Toxina del Pertussis/farmacología , Pirimidinas/farmacología , Receptores CXCR3/antagonistas & inhibidores
12.
Hepatology ; 55(5): 1610-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22237831

RESUMEN

UNLABELLED: Recent data suggest that the chemokine receptor CXCR3 is functionally involved in fibroproliferative disorders, including liver fibrosis. Neoangiogenesis is an important pathophysiological feature of liver scarring, but a functional role of angiostatic CXCR3 chemokines in this process is unclear. We therefore investigated neoangiogenesis in carbon tetrachloride (CCl(4))-induced liver fibrosis in Cxcr3(-/-) and wildtype mice by histological, molecular, and functional imaging methods. Furthermore, we assessed the direct role of vascular endothelial growth factor (VEGF) overexpression on liver angiogenesis and the fibroproliferative response using a Tet-inducible bitransgenic mouse model. The feasibility of attenuation of angiogenesis and associated liver fibrosis by therapeutic treatment with the angiostatic chemokine Cxcl9 was systematically analyzed in vitro and in vivo. The results demonstrate that fibrosis progression in Cxcr3(-/-) mice was strongly linked to enhanced neoangiogenesis and VEGF/VEGFR2 expression compared with wildtype littermates. Systemic VEGF overexpression led to a fibrogenic response within the liver and was associated with a significantly increased Cxcl9 expression. In vitro, Cxcl9 displayed strong antiproliferative and antimigratory effects on VEGF-stimulated endothelial cells and stellate cells by way of reduced VEGFR2 (KDR), phospholipase Cγ (PLCγ), and extracellular signal-regulated kinase (ERK) phosphorylation, identifying this chemokine as a direct counter-regulatory molecule of VEGF signaling within the liver. Accordingly, systemic administration of Cxcl9 led to a strong attenuation of neoangiogenesis and experimental liver fibrosis in vivo. CONCLUSION: The results identify direct angiostatic and antifibrotic effects of the Cxcr3 ligand Cxcl9 in a model of experimental liver fibrosis. The amelioration of liver damage by systemic application of Cxcl9 might offer a novel therapeutic approach for chronic liver diseases associated with increased neoangiogenesis.


Asunto(s)
Quimiocina CXCL9/farmacología , Hepatocitos/citología , Cirrosis Hepática/tratamiento farmacológico , Neovascularización Patológica/patología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Análisis de Varianza , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Quimiocina CXCL9/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Hepatocitos/metabolismo , Interferón gamma/metabolismo , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Patológica/tratamiento farmacológico , Distribución Aleatoria
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 27(12): 1288-90, 1294, 2011 Dec.
Artículo en Chino | MEDLINE | ID: mdl-22152806

RESUMEN

AIM: To construct recombinant human CXCR3B gene expression vector and obtain L929-CXCR3B gene transfected cell line for stably expressing human CXCR3B. METHODS: Human CXCR3B gene of full length was amplified by PCR from the plasmid pMD19-T/huCXCR3A. Then, it was inserted into eukaryotic expression vector pIRES2-EGFP to construct recombinant vector pIRES2-EGFP/huCXCR3B. The recombinant vector was transfected into L929 cells with Lipofectamine(TM); 2000, and cell clones stably expressing human CXCR3B molecule were screened by G418.We used FCM and RT-PCR to verify expression of CXCR3B from protein level and gene level. The ability of proliferation of L929-huCXCR3B under the circumstance of CXCR3B ligand called Mig was analyzed via MTT methods. RESULTS: We have constructed recombinant human CXCR3B gene expression vector and obtained L929-huCXCR3B gene transfected cell line which can stably express human CXCR3B molecule. The positive expression rate of CXCR3B on L929-huCXCR3B cells was 93&. The result of MTT assay showed that the proliferation of L929-huCXCR3B cells were inhibited when the cells were cocultured with Mig after 24 h, 48 h and 72 h, and the inhibition ratio were 41.44&, 44.01& and 24.80& respectively. CONCLUSION: Construction of L929-huCXCR3B cell line has laid a good foundation on research of the CXCR3B signal pathway and preparation of the CXCR3B monoclonal antibody.


Asunto(s)
Receptores CXCR4/genética , Transfección , Línea Celular , Proliferación Celular , Quimiocina CXCL9/farmacología , Clonación Molecular , Citometría de Flujo , Vectores Genéticos , Humanos , Receptores CXCR4/fisiología
14.
PLoS One ; 6(9): e25406, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21966521

RESUMEN

Adult neural precursor cells (NPCs) respond to injury or disease of the CNS by migrating to the site of damage or differentiating locally to replace lost cells. Factors that mediate this injury induced NPC response include chemokines and pro-inflammatory cytokines, such as tumor necrosis factor-α (TNFα) and interferon-γ (IFNγ), which we have shown previously promotes neuronal differentiation. RT-PCR was used to compare expression of chemokines and their receptors in normal adult mouse brain and in cultured NPCs in response to IFNγ and TNFα. Basal expression of many chemokines and their receptors was found in adult brain, predominantly in neurogenic regions, with OB≫SVZ>hippocampus and little or no expression in non-neurogenic regions, such as cortex. Treatment of SVZ-derived NPCs with IFNγ and TNFα (alone and in combination) resulted in significant upregulation of expression of specific chemokines, with CXCL1, CXCL9 and CCL2 most highly upregulated and CCL19 downregulated. Unlike IFNγ, chemokine treatment of NPCs in vitro had little or no effect on survival, proliferation or migration. Neuronal differentiation was promoted by CXCL9, CCL2 and CCL21, while astrocyte and total oligodendrocyte differentiation was not affected. However, IFNγ, CXCL1, CXCL9 and CCL2 promoted oligodendrocyte maturation. Therefore, not only do NPCs express chemokine receptors, they also produce several chemokines, particularly in response to inflammatory mediators. This suggests that autocrine or paracrine production of specific chemokines by NPCs in response to inflammatory mediators may regulate differentiation into mature neural cell types and may alter NPC responsiveness to CNS injury or disease.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Animales , Células Cultivadas , Quimiocina CCL19/metabolismo , Quimiocina CCL19/farmacología , Quimiocina CCL2/metabolismo , Quimiocina CCL2/farmacología , Quimiocina CXCL1/metabolismo , Quimiocina CXCL1/farmacología , Quimiocina CXCL9/metabolismo , Quimiocina CXCL9/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Técnicas In Vitro , Interferón gamma/farmacología , Ratones , Neuronas/metabolismo , Bulbo Olfatorio/efectos de los fármacos , Bulbo Olfatorio/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/farmacología
15.
Proc Natl Acad Sci U S A ; 108(41): 17159-64, 2011 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-21949405

RESUMEN

Chemokines are a family of chemotactic cytokines that function in host defense by orchestrating cellular movement during infection. In addition to this function, many chemokines have also been found to mediate the direct killing of a range of pathogenic microorganisms through an as-yet-undefined mechanism. As an understanding of the molecular mechanism and microbial targets of chemokine-mediated antimicrobial activity is likely to lead to the identification of unique, broad-spectrum therapeutic targets for effectively treating infection, we sought to investigate the mechanism by which the chemokine CXCL10 mediates bactericidal activity against the Gram-positive bacterium Bacillus anthracis, the causative agent of anthrax. Here, we report that disruption of the gene ftsX, which encodes the transmembrane domain of a putative ATP-binding cassette transporter, affords resistance to CXCL10-mediated antimicrobial effects against vegetative B. anthracis bacilli. Furthermore, we demonstrate that in the absence of FtsX, CXCL10 is unable to localize to its presumed site of action at the bacterial cell membrane, suggesting that chemokines interact with specific, identifiable bacterial components to mediate direct microbial killing. These findings provide unique insight into the mechanism of CXCL10-mediated bactericidal activity and establish, to our knowledge, the first description of a bacterial component critically involved in the ability of host chemokines to target and kill a bacterial pathogen. These observations also support the notion of chemokine-mediated antimicrobial activity as an important foundation for the development of innovative therapeutic strategies for treating infections caused by pathogenic, potentially multidrug-resistant microorganisms.


Asunto(s)
Bacillus anthracis/inmunología , Proteínas Bacterianas/inmunología , Proteínas de Ciclo Celular/inmunología , Quimiocinas CXC/fisiología , Animales , Antiinfecciosos/farmacología , Bacillus anthracis/efectos de los fármacos , Bacillus anthracis/genética , Bacillus anthracis/ultraestructura , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Membrana Celular/inmunología , Membrana Celular/ultraestructura , Quimiocina CXCL10/farmacología , Quimiocina CXCL10/fisiología , Quimiocina CXCL9/farmacología , Quimiocina CXCL9/fisiología , Farmacorresistencia Microbiana/genética , Eliminación de Gen , Genes Bacterianos , Prueba de Complementación Genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Ratones , Microscopía Electrónica de Transmisión , Mutación , Esporas Bacterianas/efectos de los fármacos , Esporas Bacterianas/inmunología
16.
Biomed Pharmacother ; 65(8): 547-54, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21775092

RESUMEN

Mucositis arising from cancer chemotherapy is a common problem for which there is no definitive treatment. 5-fluorouracil (5-FU) is a common cytotoxic agent used to treat solid tumors. A global gene expression array was performed to identify genetic signals involved in the pathogenesis of mucositis. The chemokine (C-X-C motif) ligand 9 (CXCL9) was one of the candidates identified that presented a characteristic gene expression profile; its temporal expression pattern was correlated with the damage and regeneration phases of the small intestine upon 5-FU chemotherapy. We found that prophylactic CXCL9 administration was able to attenuate the severity of mucositis, weight loss and diarrhea caused by chemotherapy. CXCL9 also increased the tolerance of the mice to lethal-dose chemotherapy. Moreover, we demonstrated that CXCL9 was able to promote the proliferation and regeneration of intestinal cells by inhibiting the proliferation of normal intestinal mucosal cells prior to chemotherapy and by reducing the 5-FU-induced apoptosis in intestinal crypts. Thus, pretreatment with CXCL9 is a new and promising strategy for the alleviation of chemotherapy-induced intestinal mucositis in clinical settings.


Asunto(s)
Quimiocina CXCL9/farmacología , Fluorouracilo/toxicidad , Mucosa Intestinal/efectos de los fármacos , Mucositis/prevención & control , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Antimetabolitos Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Diarrea/inducido químicamente , Diarrea/prevención & control , Femenino , Fluorouracilo/administración & dosificación , Perfilación de la Expresión Génica , Humanos , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Mucositis/inducido químicamente , Mucositis/genética , Índice de Severidad de la Enfermedad , Pérdida de Peso/efectos de los fármacos
17.
Immunology ; 132(4): 503-15, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21255008

RESUMEN

The chemokine receptor CXCR3 and its ligands CXCL9, CXCL10 and CXCL11 are involved in variety of inflammatory disorders including multiple sclerosis, rheumatoid arthritis, psoriasis and sarcoidosis. Two alternatively spliced variants of the human CXCR3-A receptor have been described, termed CXCR3-B and CXCR3-alt. Human CXCR3-B binds CXCL9, CXCL10, CXCL11 as well as an additional ligand CXCL4. In contrast, CXCR3-alt only binds CXCL11. We report that CXCL4 induces intracellular calcium mobilization as well as Akt and p44/p42 extracellular signal-regulated kinase phosphorylation, in activated human T lymphocytes. These responses have similar concentration dependence and time-courses to those induced by established CXCR3 agonists. Moreover, phosphorylation of Akt and p44/p42 is inhibited by pertussis toxin, suggesting coupling to Gα(i) protein. Surprisingly, and in contrast with the other CXCR3 agonists, stimulation of T lymphocytes with CXCL4 failed to elicit migratory responses and did not lead to loss of surface CXCR3 expression. Taken together, our findings show that, although CXCL4 is coupled to downstream biochemical machinery, its role in T cells is probably distinct from that of CXCR3-A agonists.


Asunto(s)
Variación Genética , Receptores CXCR3/genética , Linfocitos T/metabolismo , Acetamidas/farmacología , Western Blotting , Calcio/metabolismo , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocina CXCL10/farmacología , Quimiocina CXCL11/genética , Quimiocina CXCL11/farmacología , Quimiocina CXCL9/genética , Quimiocina CXCL9/farmacología , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Toxina del Pertussis/farmacología , Fosforilación/efectos de los fármacos , Factor Plaquetario 4/genética , Factor Plaquetario 4/farmacología , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Receptores CXCR3/agonistas , Receptores CXCR3/metabolismo , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/citología , Linfocitos T/efectos de los fármacos
18.
J Biol Chem ; 286(7): 5069-77, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21138841

RESUMEN

There are few examples of host signals that are beneficial to bacteria during infection. Here we found that 31 out of 42 host immunoregulatory chemokines were able to induce release of the virulence factor protein A (SPA) from a strain of community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA). Detailed study of chemokine CXCL9 revealed that SPA release occurred through a post-translational mechanism and was inversely proportional to bacterial density. CXCL9 bound specifically to the cell membrane of CA-MRSA, and the related SPA-releasing chemokine CXCL10 bound to both cell wall and cell membrane. Clinical samples from patients infected with S. aureus and samples from a mouse model of CA-MRSA skin abscess all contained extracellular SPA. Further, SPA-releasing chemokines were present in mouse skin lesions infected with CA-MRSA. Our data identify a potential new mode of immune evasion, in which the pathogen exploits a host defense factor to release a virulence factor; moreover, chemokine binding may serve a scavenging function in immune evasion by S. aureus.


Asunto(s)
Pared Celular/inmunología , Quimiocina CXCL9/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Proteína Estafilocócica A/inmunología , Staphylococcus aureus/inmunología , Factores de Virulencia/inmunología , Animales , Membrana Celular/inmunología , Membrana Celular/metabolismo , Pared Celular/metabolismo , Quimiocina CXCL9/metabolismo , Quimiocina CXCL9/farmacología , Humanos , Ratones , Unión Proteica/inmunología , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Factores de Virulencia/metabolismo
19.
J Biol Chem ; 284(43): 29499-508, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19628464

RESUMEN

The anaerobic bacterium Finegoldia magna is part of the human commensal microbiota, but is also an important opportunistic pathogen. This bacterium expresses a subtilisin-like serine proteinase, SufA, which partially degrade the antibacterial chemokine MIG/CXCL9. Here, we show that MIG/CXCL9 is produced by human keratinocytes in response to inflammatory stimuli. In contrast to the virulent human pathogen Streptococcus pyogenes, the presence of F. magna had no enhancing effect on the MIG/CXCL9 expression by keratinocytes, suggesting poor detection of the latter by pathogen-recognition receptors. When MIG/CXCL9 was exposed to SufA-expressing F. magna, the molecule was processed into several smaller fragments. Analysis by mass spectrometry showed that SufA cleaves MIG/CXCL9 at several sites in the COOH-terminal region of the molecule. At equimolar concentrations, SufA-generated MIG/CXCL9 fragments were not bactericidal against F. magna, but retained their ability to kill S. pyogenes. Moreover, the SufA-generated MIG/CXCL9 fragments were capable of activating the angiostasis-mediating CXCR3 receptor, which is expressed on endothelial cells, in an order of magnitude similar to that of intact MIG/CXCL9. F. magna expresses a surface protein called FAF that is released from the bacterial surface by SufA. Soluble FAF was found to bind and inactivate the antibacterial activity of MIG/CXCL9, thereby further potentially promoting the survival of F. magna. The findings suggest that SufA modulation of the inflammatory response could be a mechanism playing an important role in creating an ecologic niche for F. magna, decreasing antibacterial activity and suppressing angiogenesis, thus providing advantage in survival for this anaerobic opportunist compared with competing pathogens during inflammation.


Asunto(s)
Antibacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Quimiocina CXCL9/metabolismo , Bacilos Grampositivos Formadores de Endosporas/metabolismo , Queratinocitos/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Antibacterianos/farmacología , Línea Celular , Quimiocina CXCL9/farmacología , Infecciones por Bacterias Grampositivas/metabolismo , Humanos , Inflamación/metabolismo , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Receptores CXCR3/agonistas , Receptores CXCR3/metabolismo , Streptococcus pyogenes/metabolismo
20.
J Pharmacol Exp Ther ; 329(3): 1142-7, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19258523

RESUMEN

A constitutively active G protein-coupled receptor (GPCR) encoded by Kaposi's sarcoma-associated herpesvirus (human herpesvirus-8) (KSHV) is expressed in endothelial (spindle) cells of Kaposi's sarcoma lesions. In this study, we report novel effects of basal signaling by this receptor and of inverse agonist chemokines on migration of KSHV-GPCR-expressing mouse lung endothelial cells. We show that basal signaling by KSHV-GPCR inhibits migration of endothelial cells in two systems, movement through porous filters and in vitro wound closure. Naturally occurring chemokines, interferon gamma-inducible protein-10 and stromal-derived factor-1, which act as inverse agonists at KSHV-GPCR, abrogate the inhibition of migration and stimulate directed migration (or chemotaxis) of these cells. Thus, the expression of KSHV-GPCR may allow infected endothelial cells in situ to remain in a localized environment or to directionally migrate along a gradient of specific chemokines that are inverse agonists at KSHV-GPCR.


Asunto(s)
Quimiocinas/farmacología , Quimiotaxis/fisiología , Agonismo Inverso de Drogas , Células Endoteliales/citología , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/metabolismo , Transducción de Señal/fisiología , Animales , Línea Celular , Quimiocina CXCL1/farmacología , Quimiocina CXCL10/farmacología , Quimiocina CXCL11/farmacología , Quimiocina CXCL12/farmacología , Quimiocina CXCL9/farmacología , Quimiotaxis/efectos de los fármacos , Células Endoteliales/metabolismo , Ratones , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Receptores de Quimiocina/agonistas , Transducción de Señal/efectos de los fármacos , Transfección , Cicatrización de Heridas/efectos de los fármacos , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...