Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Clin. transl. oncol. (Print) ; 25(1): 160-172, ene. 2023.
Article in English | IBECS | ID: ibc-215830

ABSTRACT

Purpose Colorectal cancer (CRC) is a malignant tumor. Oxaliplatin (OXA) can inhibit cancer-associated fibroblasts (CAFs)-induced cancer progression. This study sought to explore the mechanism of OXA in CAFs-induced CRC development. Methods CRC cell lines (Caco-2, SW620), normal fibroblasts (NFs), and CAFs were treated with OXA. NFs and CAFs were cultured. CAFs were treated with/without OXA (0.4 mM), and the supernatant was extracted as the conditioned medium (CM) to culture CRC cells. Cell malignant episodes, E-cadherin and Vimentin levels, CXCL1, CXCL2, CXCL3, CXCL8, and CXCL11 mRNA levels, CXCL11 protein level, and extracellular release were assessed. CAFs were transfected with interfering RNA sh-CXCL11 to silence CXCL11 or transfected with CXCL11 overexpression plasmids and treated with OXA to explore the role of CXCL11 in OXA-mediated CRC cells through CAFs. CXCL11 receptor CXCR3 levels in CRC cells and the PI3K/AKT pathway changes were examined. The xenogeneic tumor was transplanted in nude mice. CXCL11 and CXCR3 levels in tumor tissues, tumor volume, shape, size, weight, and Ki67 positive expressions were assessed. Results CRC cell growths and epithelial–mesenchymal transformation were stimulated after culture with CAFs–CM, while OXA averted these trends. CXCL11 mRNA level was elevated most significantly, and its protein and extracellular secretion levels were raised, while OXA diminished the levels. CXCL11 silencing weakened the effects of CAFs–CM on promoting CRC proliferation and malignant episodes and CXCL11 overexpression averted OXA property on inhibiting CAFs-promoted CRC cell growth (AU)


Subject(s)
Animals , Mice , Cancer-Associated Fibroblasts , Colorectal Neoplasms/metabolism , Oxaliplatin/pharmacology , Antineoplastic Agents/pharmacology , Caco-2 Cells , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Chemokine CXCL11/metabolism , Chemokine CXCL11/pharmacology , Mice, Nude , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt , Receptors, CXCR3/metabolism
2.
Clin Transl Oncol ; 25(1): 160-172, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36129606

ABSTRACT

PURPOSE: Colorectal cancer (CRC) is a malignant tumor. Oxaliplatin (OXA) can inhibit cancer-associated fibroblasts (CAFs)-induced cancer progression. This study sought to explore the mechanism of OXA in CAFs-induced CRC development. METHODS: CRC cell lines (Caco-2, SW620), normal fibroblasts (NFs), and CAFs were treated with OXA. NFs and CAFs were cultured. CAFs were treated with/without OXA (0.4 mM), and the supernatant was extracted as the conditioned medium (CM) to culture CRC cells. Cell malignant episodes, E-cadherin and Vimentin levels, CXCL1, CXCL2, CXCL3, CXCL8, and CXCL11 mRNA levels, CXCL11 protein level, and extracellular release were assessed. CAFs were transfected with interfering RNA sh-CXCL11 to silence CXCL11 or transfected with CXCL11 overexpression plasmids and treated with OXA to explore the role of CXCL11 in OXA-mediated CRC cells through CAFs. CXCL11 receptor CXCR3 levels in CRC cells and the PI3K/AKT pathway changes were examined. The xenogeneic tumor was transplanted in nude mice. CXCL11 and CXCR3 levels in tumor tissues, tumor volume, shape, size, weight, and Ki67 positive expressions were assessed. RESULTS: CRC cell growths and epithelial-mesenchymal transformation were stimulated after culture with CAFs-CM, while OXA averted these trends. CXCL11 mRNA level was elevated most significantly, and its protein and extracellular secretion levels were raised, while OXA diminished the levels. CXCL11 silencing weakened the effects of CAFs-CM on promoting CRC proliferation and malignant episodes and CXCL11 overexpression averted OXA property on inhibiting CAFs-promoted CRC cell growth. CXCR3 and PI3K and AKT1 phosphorylation levels were raised in the CAFs-CM group but diminished by OXA. CXCL11 overexpression in CAFs averted OXA property on inhibiting CAFs-activated CXCR3/PI3K/AKT in CRC cells. OXA also inhibited the progression of xenograft tumors by limiting CAFs-secreted CXCL11. CONCLUSIONS: OXA repressed CRC progression by inhibiting CAFs-secreted CXCL11 and the CXCR3/PI3K/AKT pathway.


Subject(s)
Cancer-Associated Fibroblasts , Colorectal Neoplasms , Animals , Mice , Humans , Oxaliplatin/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Mice, Nude , Caco-2 Cells , Cell Line, Tumor , Fibroblasts/metabolism , Colorectal Neoplasms/genetics , Cell Proliferation , Cell Movement/genetics , Chemokine CXCL11/metabolism , Chemokine CXCL11/pharmacology , Receptors, CXCR3/metabolism
3.
Br J Dermatol ; 176(1): 127-137, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27436825

ABSTRACT

BACKGROUND: Cell migration plays a major role in the immune response and in tumorigenesis. Interferon-inducible T-cell alpha chemoattractant (ITAC) elicits a strong chemotactic response from immune cells. OBJECTIVES: To examine the effect of ITAC on melanocyte migration and pigmentation and its involvement in related disorders, and to investigate potential key players in these processes. METHODS: Human melanocytes or melanoma cells were treated with ITAC and a migration assay was carried out. Global gene expression analysis was performed to find genes regulated by ITAC treatment. The function of key players involved in ITAC-induced cellular processes was addressed using knockdown or overexpression experiments in combination with ITAC treatment. ITAC expression in the inflammation-associated hypopigmentary disorder, vitiligo, was examined. RESULTS: Among CXCR3 ligands, only ITAC induced melanocyte migration. ITAC treatment upregulated the expression of histone deacetylase 5 (HDAC5) and downregulated that of p53, a known target of HDAC5. Through knockdown or overexpression of HDAC5 and p53, we confirmed that HDAC5 mediates ITAC-induced migration by decreasing levels of p53 via deacetylation. In addition, ITAC treatment could decrease pigmentation in a p53- and HDAC5-dependent manner. Finally, the increased migration of human melanoma cells by ITAC treatment and the increased ITAC expression in the epidermis of vitiligo skin were verified. CONCLUSIONS: This study provides in vitro evidence for the migratory and hypopigmentation effects of ITAC on melanocytic cells, gives translational insights into the roles of ITAC in pathological conditions, and suggests that HDAC5 and its substrate p53 are potent targets for regulating ITAC-induced cellular processes.


Subject(s)
Cell Movement/drug effects , Chemokine CXCL11/pharmacology , Histone Deacetylases/metabolism , Hypopigmentation/enzymology , Melanocytes/drug effects , Cells, Cultured , Down-Regulation/physiology , Epidermal Cells , Gene Knockdown Techniques , Histone Deacetylases/deficiency , Humans , RNA, Messenger/metabolism , Receptors, CXCR/metabolism , Repressor Proteins/deficiency , Tumor Suppressor Protein p53/metabolism , Up-Regulation/physiology
5.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 31(11): 1544-8, 2015 Nov.
Article in Chinese | MEDLINE | ID: mdl-26522366

ABSTRACT

OBJECTIVE: To study the effects of chemokine (C-X-C motif) receptor 3 (CXCR3) monoclonal antibody (mAb) on the proliferation and migration of MCF-7 and HepG2 cells in vitro. METHODS: Ascites of CXCR3 mAb was prepared at first. MCF-7 and HepG2 cells with high expressions of CXCR3 were screened by flow cytometry. MTT assay was used to detect the effects of CXCR3 mAb on the proliferation of MCF-7 and HepG2 cells in vitro in the absence/presence of interferon-inducible T-cell alpha chemoattractant (I-TAC). Transwell™ assay was performed to investigate the effects of CXCR3 mAb on the migration of MCF-7 and HepG2 cells in vitro in the absence/presence of I-TAC. RESULTS: The expression rate of CXCR3 on MCF-7 and HepG2 cells were 83.5% and 96.2%, respectively. 50 mg/mL CXCR3 mAb significantly inhibited the proliferation and migration of MCF-7 and HepG2 cells, and also inhibited the promoting effect of I-TAC on the proliferation and migration of MCF-7 and HepG2 cells in vitro. CONCLUSION: CXCR3 mAb can significantly inhibit the proliferation and migration of the tumor cells highly expressing CXCR3 in vitro.


Subject(s)
Antibodies, Monoclonal/immunology , Receptors, CXCR3/physiology , Animals , Cell Movement , Cell Proliferation , Chemokine CXCL11/pharmacology , Hep G2 Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C
6.
J Clin Invest ; 124(5): 2009-22, 2014 05.
Article in English | MEDLINE | ID: mdl-24713654

ABSTRACT

A single G protein-coupled receptor (GPCR) can activate multiple signaling cascades based on the binding of different ligands. The biological relevance of this feature in immune regulation has not been evaluated. The chemokine-binding GPCR CXCR3 is preferentially expressed on CD4+ T cells, and canonically binds 3 structurally related chemokines: CXCL9, CXCL10, and CXCL11. Here we have shown that CXCL10/CXCR3 interactions drive effector Th1 polarization via STAT1, STAT4, and STAT5 phosphorylation, while CXCL11/CXCR3 binding induces an immunotolerizing state that is characterized by IL-10(hi) (Tr1) and IL-4(hi) (Th2) cells, mediated via p70 kinase/mTOR in STAT3- and STAT6-dependent pathways. CXCL11 binds CXCR3 with a higher affinity than CXCL10, suggesting that CXCL11 has the potential to restrain inflammatory autoimmunity. We generated a CXCL11-Ig fusion molecule and evaluated its use in the EAE model of inflammatory autoimmune disease. Administration of CXCL11-Ig during the first episode of relapsing EAE in SJL/J mice not only led to rapid remission, but also prevented subsequent relapse. Using GFP-expressing effector CD4+ T cells, we observed that successful therapy was associated with reduced accumulation of these cells at the autoimmune site. Finally, we showed that very low doses of CXCL11 rapidly suppress signs of EAE in C57BL/6 mice lacking functional CXCL11.


Subject(s)
Chemokine CXCL11/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Chemokine CXCL11/genetics , Chemokine CXCL11/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Mice , Mice, Knockout , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , STAT Transcription Factors/genetics , STAT Transcription Factors/immunology , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/pathology
7.
Mol Cancer Res ; 12(1): 38-47, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24025971

ABSTRACT

UNLABELLED: Chemokines have been implicated as key contributors of non-small cell lung cancer (NSCLC) metastasis. However, the role of CXCR7, a recently discovered receptor for CXCL12 ligand, in the pathogenesis of NSCLC is unknown. To define the relative contribution of chemokine receptors to migration and metastasis, we generated human lung A549 and H157 cell lines with stable knockdown of CXCR4, CXCR7, or both. Cancer cells exhibited chemotaxis to CXCL12 that was enhanced under hypoxic conditions, associated with a parallel induction of CXCR4, but not CXCR7. Interestingly, neither knockdown cell line differed in the rate of proliferation, apoptosis, or cell adherence; however, in both cell lines, CXCL12-induced migration was abolished when CXCR4 signaling was abrogated. In contrast, inhibition of CXCR7 signaling did not alter cellular migration to CXCL12. In an in vivo heterotropic xenograft model using A549 cells, expression of CXCR4, but not CXCR7, on cancer cells was necessary for the development of metastases. In addition, cancer cells knocked down for CXCR4 (or both CXCR4 and CXCR7) produced larger and more vascular tumors as compared with wild-type or CXCR7 knockdown tumors, an effect that was attributable to cancer cell-derived CXCR4 out competing endothelial cells for available CXCL12 in the tumor microenvironment. These results indicate that CXCR4, not CXCR7, expression engages CXCL12 to mediate NSCLC metastatic behavior. IMPLICATIONS: Targeting CXCR4-mediated migration and metastasis may be a viable therapeutic option in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Chemokine CXCL12/pharmacology , Lung Neoplasms/pathology , Neoplasm Metastasis/genetics , Receptors, CXCR4/genetics , Animals , Apoptosis/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Cell Proliferation , Chemokine CXCL11/pharmacology , Chemotaxis/genetics , Endothelial Cells/pathology , Female , Humans , Mice , Mice, SCID , RNA Interference , RNA, Small Interfering , Receptors, CXCR/genetics , Tumor Microenvironment
8.
PLoS One ; 8(10): e79610, 2013.
Article in English | MEDLINE | ID: mdl-24205388

ABSTRACT

Engineered biomatrices offer the potential to recapitulate the regenerative microenvironment, with important implications in tissue repair. In this context, investigation of the molecular interactions occurring between growth factors, cytokines and extracellular matrix (ECM) has gained increasing interest. Here, we sought to investigate the possible interactions between the ECM proteins fibronectin (FN) and fibrinogen (Fg) with the CXCR3 ligands CXCL9, CXCL10 and CXCL11, which are expressed during wound healing. New binding interactions were observed and characterized. Heparin-binding domains within Fg (residues 15-66 of the ß chain, Fg ß15-66) and FN (FNI1-5, but not FNIII12-14) were involved in binding to CXCL10 and CXCL11 but not CXCL9. To investigate a possible influence of FN and Fg interactions with CXCL11 in mediating its role during re-epithelialization, we investigated human keratinocyte migration in vitro and wound healing in vivo in diabetic db/db mice. A synergistic effect on CXCL11-induced keratinocyte migration was observed when cells were treated with CXCL11 in combination with FN in a transmigration assay. Moreover, wound healing was enhanced in full thickness excisional wounds treated with fibrin matrices functionalized with FN and containing CXCL11. These findings highlight the importance of the interactions occurring between cytokines and ECM and point to design concepts to develop functional matrices for regenerative medicine.


Subject(s)
Fibrinogen/physiology , Fibronectins/physiology , Wound Healing/physiology , Animals , Cell Movement/drug effects , Chemokine CXCL10/metabolism , Chemokine CXCL11/metabolism , Chemokine CXCL11/pharmacology , Chemokine CXCL9/metabolism , Extracellular Matrix/metabolism , Fibrinogen/metabolism , Fibronectins/metabolism , Fibronectins/pharmacology , HEK293 Cells , Heparin/pharmacology , Humans , Keratin-16/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/physiology , Male , Mice, Inbred C57BL , Receptors, CXCR3/metabolism , Regenerative Medicine
9.
Cell Signal ; 25(4): 729-35, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23277202

ABSTRACT

CC and CXC chemokine receptor signalling networks are regulated in different ways. Here we show that intracellular calcium release and cell migration occur independent of Gßγ activation in response to CCL3, whereas CXCL11 induced migration of activated T-lymphocytes depends on Gßγ activation. Treatment of a range of cell types with gallein, a pharmacological inhibitor of Gßγ signalling, did not result in a reduction in CCL3 induced cellular migration, but resulted in enhanced calcium mobilisation following chemokine stimulation. Inhibition of PI3 kinase (PI3K) and AKT, which are activated downstream of Gßγ, equally had no effect on calcium release and a minor effect on cell migration. Similarly, inhibition of ERK1/2 did not prevent CCL3 induced migration. Interestingly, Gßγ as well as PI3K activation is necessary for CXCL11 induced migration of activated T-cells. These data not only confirm a role for Gßγ signalling in CXCL11 induced migration, but also demonstrate that targeting Gßγ as a therapeutic target to prevent migration in inflammatory disease may not be beneficial, at least not for CCL3 induced migration. This highlights the distinct differences in the mechanisms on how CC- and CXC-receptors activate cellular migration.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Receptors, Chemokine/metabolism , Animals , CHO Cells , Calcium/metabolism , Cell Line , Cell Movement/drug effects , Chemokine CCL3/pharmacology , Chemokine CXCL11/pharmacology , Chemotaxis/drug effects , Cricetinae , Cricetulus , HEK293 Cells , HeLa Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CCR5/metabolism , Signal Transduction/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xanthenes/pharmacology
10.
Biochem Biophys Res Commun ; 419(2): 412-8, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22349504

ABSTRACT

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.


Subject(s)
Electric Impedance , GTP-Binding Protein alpha Subunits, Gi-Go/agonists , Isoquinolines/pharmacology , Receptors, CXCR3/agonists , Acetamides/pharmacology , Chemokine CXCL10/pharmacology , Chemokine CXCL11/pharmacology , Chemokine CXCL9/pharmacology , HEK293 Cells , Humans , Ligands , Metabolic Networks and Pathways , Pertussis Toxin/pharmacology , Pyrimidines/pharmacology , Receptors, CXCR3/antagonists & inhibitors
11.
J Immunol ; 186(8): 4936-45, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21402888

ABSTRACT

SHIP-1 negatively regulates the PI3K pathway in hematopoietic cells and has an emerging role in T lymphocyte biology. PI3K and SHIP can regulate cell migration in leukocytes, particularly in neutrophils, although their role in T cell migration has been less clear. Therefore, we sought to explore the role of SHIP-1 in human CD4(+) T lymphocyte cell migration responses to chemoattractants using a lentiviral-mediated expression system and a short hairpin RNA approach. Silencing of SHIP-1 leads to increased basal phosphorylation of protein kinase B/Akt and its substrate GSK3ß, as well as an increase in basal levels of polymerized actin, suggesting that SHIP-1 might regulate changes in the cytoskeleton. Accordingly, silencing of SHIP-1 led to loss of microvilli and ezrin/radixin/moesin phosphorylation, which could not be rescued by the PI3K inhibitor Ly294002. There were striking morphological changes, including a loss of microvilli projections, which mirrored changes in wild type cells after stimulation with the chemokine CXCL11. There was no defect in directional T cell migration toward CXCL11 in the SHIP-1-silenced cells but, importantly, there was a defect in the overall basal motility of SHIP-1 knockdown cells. Taken together, these results implicate SHIP-1 as a key regulator of basal PI3K signaling in human CD4(+) T lymphocytes with important phosphatase-independent actions, which together are key for maintaining normal morphology and basal motility.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Cell Movement , Phosphoric Monoester Hydrolases/metabolism , Actins/metabolism , Blotting, Western , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/ultrastructure , Cell Survival , Cells, Cultured , Chemokine CXCL11/pharmacology , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Flow Cytometry , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Inositol Polyphosphate 5-Phosphatases , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Microscopy, Electron, Scanning , Microvilli/metabolism , Microvilli/ultrastructure , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction
12.
Immunology ; 132(4): 503-15, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21255008

ABSTRACT

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.


Subject(s)
Genetic Variation , Receptors, CXCR3/genetics , T-Lymphocytes/metabolism , Acetamides/pharmacology , Blotting, Western , Calcium/metabolism , Cell Movement/drug effects , Cells, Cultured , Chemokine CXCL10/genetics , Chemokine CXCL10/pharmacology , Chemokine CXCL11/genetics , Chemokine CXCL11/pharmacology , Chemokine CXCL9/genetics , Chemokine CXCL9/pharmacology , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression/drug effects , HEK293 Cells , Humans , Pertussis Toxin/pharmacology , Phosphorylation/drug effects , Platelet Factor 4/genetics , Platelet Factor 4/pharmacology , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Receptors, CXCR3/agonists , Receptors, CXCR3/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
13.
Eur J Haematol ; 85(6): 472-83, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20887389

ABSTRACT

CXCR7 was identified as another stromal-derived factor-1 (SDF-1)-binding receptor that also binds the interferon-inducible T-cell chemoattractant (I-TAC), and we became interested in its potential role in migration/adhesion of normal hematopoietic stem/progenitor cells (HSPCs) as well as selected leukemia cell lines. To address this normal human bone marrow-, umbilical cord blood-, and mobilized peripheral blood-derived cells as well as 16 selected human leukemic cell lines were phenotyped for CXCR7 expression. The expression of CXCR7 in hematopoietic cell lines was analyzed at transcriptional level. The biologic significance of CXCR7 expression was subsequently tested in signal transduction studies as well as in in vitro proliferation and chemotactic assays. We noted that CXCR7 is expressed at very low levels (approximately 3-6%) in normal human CD34(+) cells isolated from bone marrow, umbilical cord blood, and mobilized peripheral blood. More importantly, when we employed I-TAC, which activates CXCR7, but not CXCR4, we did not observe any chemotactic responsiveness in human clonogenic progenitors. As expected, I-TAC also did not affect clonogenic growth of human CD34(+) cells. In contrast, functional CXCR7, whose expression is regulated in an NF-κΒ-dependent manner, as we report here, is highly expressed in several human myeloid malignant cell lines. I-TAC-induced activation of CXCR7 in human hematopoietic cell lines leads to phosphorylation of MAPKp42/44 and AKT, and enhanced cell adhesion and slightly cell migration. In conclusion, CXCR7 is expressed at very low level on normal human HSPCs and does not play a direct role in their proliferation or slightly cell migration; however, in contrast, it is involved in trafficking/adhesion of human leukemic cells.


Subject(s)
Antigens, CD34 , Chemokine CXCL12/metabolism , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Receptors, CXCR/biosynthesis , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Chemokine CXCL11/pharmacology , Chemokine CXCL12/genetics , Fetal Blood/metabolism , HL-60 Cells , Hematopoietic Stem Cells/pathology , Humans , Jurkat Cells , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplastic Stem Cells/pathology , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , U937 Cells
14.
Vet Immunol Immunopathol ; 136(1-2): 170-5, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20347492

ABSTRACT

This report describes the cloning and characterization of expressed gene sequences of the swine and bovine interferon-gamma inducible chemokine CXCL11, or I-TAC, associated with type 1 T-helper immune responses, and affirmation of bioactivity of their yeast-expressed protein products. The coding regions of both cDNA sequences were 303 nucleotides in length; each is coded for four exons in the genome. The bovine coding region shared 82% and 70% homology with human and mouse CXCL11, respectively, and the swine coding region 84% and 72% homology, respectively. As expected the swine and bovine CXCL11 sequences showed less homology with other human and mouse C-X-C motif chemokine sequences. Each cDNA was cloned into plasmids and transfected into Pichia pastoris (yeast) and the resultant expressed protein purified. Biological activity of each purified chemokine was affirmed by chemotaxis assays. Both swine and bovine CXCL11 were chemotactic for mitogen and IL-2 stimulated peripheral blood mononuclear cells. This is the first report for bioactivity of this chemokine in livestock species. This work provides valuable new reagents for investigating basic immunity as well as vaccine and disease responses in swine and cattle, goals of the U.S. Veterinary Immune Reagent Network which supported this effort.


Subject(s)
Cattle/genetics , Cattle/immunology , Chemokine CXCL11/genetics , Sus scrofa/genetics , Sus scrofa/immunology , Amino Acid Sequence , Animals , Base Sequence , Chemokine CXCL11/pharmacology , Chemotaxis, Leukocyte/drug effects , Cloning, Molecular , DNA Primers/genetics , Gene Expression , Humans , Interferon-gamma/pharmacology , Mice , Molecular Sequence Data , Phylogeny , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Th1 Cells/immunology
15.
Connect Tissue Res ; 51(2): 113-22, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20001843

ABSTRACT

Autologous human serum is used in cartilage repair and may exert its effect by the recruitment of mesenchymal stem and progenitor cells (MSC). Aim of our study was to analyze the chemokine profile of human serum and to verify chemotactic activity of selected chemokines on MSC. Human MSC were isolated from iliac crest bone marrow aspirates. Chemotactic activity of human serum made from whole blood and pharma grade serum was tested in 96-well chemotaxis assays and chemokine levels were analyzed using human chemokine antibody membrane arrays. The chemotactic potential of selected chemokines on MSC was tested dose dependently using chemotaxis assays. Human serum derived from whole blood significantly attracted human MSC, while pharma grade serum did not recruit MSC. Human chemokine antibody array analysis showed that the level of chemokines CXCL-3, 5, 7-8, 10-12, 16; CCL- 2, 5, 11, 13, 16-20, 24-25, 27; as well as XCL-1 was elevated (fold change >1.5) in serum derived from whole blood compared to nonrecruiting pharma grade serum. Chemotaxis assays showed that the chemokines IP-10/CXCL-10 and I-TAC/CXCL-11 significantly recruit human MSC. PARC/CCL-18, HCC-4/CCL-16, CTACK/CCL-27, and Lymphotactin/XCL-1 showed no chemotactic effect on MSC. Therefore, human serum derived from whole blood contains chemokines that may contribute to serum-mediated recruitment of human mesenchymal progenitors from bone marrow.


Subject(s)
Chemokine CXCL10/pharmacology , Chemokine CXCL11/pharmacology , Chemokines/blood , Chemotaxis/drug effects , Mesenchymal Stem Cells/drug effects , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Cell Separation , Cells, Cultured , Chemokine CXCL10/blood , Chemokine CXCL11/blood , Female , Flow Cytometry , Humans , Immunoenzyme Techniques , Male , Mesenchymal Stem Cells/physiology , Middle Aged
16.
Methods Enzymol ; 461: 397-412, 2009.
Article in English | MEDLINE | ID: mdl-19480929

ABSTRACT

The ability of chemokines to induce the migration of cells expressing their cognate G-protein-coupled receptor is a characteristic property of chemokine function. To study this important function, in vitro chemotaxis assays are most often used, which, although useful, lack many components of the complex in vivo trafficking process. Reliable in vivo recruitment assays have been very difficult to establish. We describe a robust in vivo T-cell recruitment assay for adoptively transferred T lymphocytes in mice. Instillation of the CXCR3 chemokine ligands IP-10/CXCL10 or I-TAC/CXCL11 into the airways results in robust recruitment of transferred T lymphocytes. The assay thereby models the natural environment of chemokine function, as chemokines are expressed in the airways during inflammation, inducing selective leukocyte homing. This assay is particularly useful for the analysis of chemokine and chemokine receptor mutants in structure function studies and for testing the in vivo efficacy of inhibitory chemokine and chemokine receptor antibodies and small molecule antagonists.


Subject(s)
Chemokines/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Cell Movement/drug effects , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL10/pharmacology , Chemokine CXCL11/genetics , Chemokine CXCL11/metabolism , Chemokine CXCL11/pharmacology , Chemokines/genetics , Chemokines/metabolism , Mice , Receptors, Chemokine/genetics , Receptors, Chemokine/physiology
17.
J Pharmacol Exp Ther ; 329(3): 1142-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19258523

ABSTRACT

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.


Subject(s)
Chemokines/pharmacology , Chemotaxis/physiology , Drug Inverse Agonism , Endothelial Cells/cytology , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/metabolism , Signal Transduction/physiology , Animals , Cell Line , Chemokine CXCL1/pharmacology , Chemokine CXCL10/pharmacology , Chemokine CXCL11/pharmacology , Chemokine CXCL12/pharmacology , Chemokine CXCL9/pharmacology , Chemotaxis/drug effects , Endothelial Cells/metabolism , Mice , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Phosphorylation/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Receptors, Chemokine/agonists , Signal Transduction/drug effects , Transfection , Wound Healing/drug effects , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/metabolism
18.
Infect Immun ; 77(4): 1664-78, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19179419

ABSTRACT

Based on previous studies showing that host chemokines exert antimicrobial activities against bacteria, we sought to determine whether the interferon-inducible Glu-Leu-Arg-negative CXC chemokines CXCL9, CXCL10, and CXCL11 exhibit antimicrobial activities against Bacillus anthracis. In vitro analysis demonstrated that all three CXC chemokines exerted direct antimicrobial effects against B. anthracis spores and bacilli including marked reductions in spore and bacillus viability as determined using a fluorometric assay of bacterial viability and CFU determinations. Electron microscopy studies revealed that CXCL10-treated spores failed to undergo germination as judged by an absence of cytological changes in spore structure that occur during the process of germination. Immunogold labeling of CXCL10-treated spores demonstrated that the chemokine was located internal to the exosporium in association primarily with the spore coat and its interface with the cortex. To begin examining the potential biological relevance of chemokine-mediated antimicrobial activity, we used a murine model of inhalational anthrax. Upon spore challenge, the lungs of C57BL/6 mice (resistant to inhalational B. anthracis infection) had significantly higher levels of CXCL9, CXCL10, and CXCL11 than did the lungs of A/J mice (highly susceptible to infection). Increased CXC chemokine levels were associated with significantly reduced levels of spore germination within the lungs as determined by in vivo imaging. Taken together, our data demonstrate a novel antimicrobial role for host chemokines against B. anthracis that provides unique insight into host defense against inhalational anthrax; these data also support the notion for an innovative approach in treating B. anthracis infection as well as infections caused by other spore-forming organisms.


Subject(s)
Anti-Bacterial Agents , Bacillus anthracis/drug effects , Chemokines, CXC , Interferons/immunology , Spores, Bacterial/drug effects , Animals , Anthrax/immunology , Anthrax/microbiology , Anti-Bacterial Agents/immunology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacillus anthracis/pathogenicity , Bacillus anthracis/physiology , Chemokine CXCL10/immunology , Chemokine CXCL10/pharmacology , Chemokine CXCL10/therapeutic use , Chemokine CXCL11/immunology , Chemokine CXCL11/pharmacology , Chemokine CXCL11/therapeutic use , Chemokine CXCL9/immunology , Chemokine CXCL9/pharmacology , Chemokine CXCL9/therapeutic use , Chemokines, CXC/immunology , Chemokines, CXC/pharmacology , Chemokines, CXC/therapeutic use , Colony Count, Microbial , Female , Humans , Lung/immunology , Lung/microbiology , Mice , Mice, Inbred C57BL , Spores, Bacterial/pathogenicity
19.
J Interferon Cytokine Res ; 28(12): 733-40, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18937547

ABSTRACT

Interferons (IFNs) have proven antitumor activity against a variety of human malignancies, which may result, at least in part, from inhibition of angiogenesis. The objective of this study was to identify IFN-stimulated genes (ISGs) that played a role in mediation of angiogenic inhibition. IFN-beta was a more potent antiangiogenic agent compared to IFN-alpha2b (80% versus 20%, respectively) and suggests that IFNs inhibited angiogenesis by preventing endothelial cell differentiation, and not by direct antiproliferative effects. To identify ISGs that were key inhibitors of angiogenesis, we utilized an in vitro fibrin gel angiogenic assay which closely recapitulated the in vivo processes of angiogenesis. DNA microarray analysis of IFN-beta-treated endothelial cells in the fibrin gel assay identified 11 ISGs that were induced >10-fold during angiogenesis inhibition. Recombinant IP-10 inhibited angiogenesis in a dose-dependent fashion, but was a less effective inhibitor compared to IFN-beta, suggesting that additional ISGs are involved in inhibiting angiogenesis. ISG20 was upregulated by microarray analysis, but did not inhibit angiogenesis when overexpressed in human umbilical vein endothelial cells (HUVECs). However, a dominant negative mutant of ISG20 inhibited angiogenesis by 43%. Results suggest that IFN-induced angiogenic inhibition was likely mediated by multiple ISGs; our novel finding is that decreased exonuclease activity in HUVECs associated with expression of the ISG20 ExoII mutant inhibited angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Immunologic Factors/pharmacology , Interferon-beta/pharmacology , Neovascularization, Pathologic/immunology , Neovascularization, Physiologic/drug effects , Animals , Chemokine CXCL10/pharmacology , Chemokine CXCL11/pharmacology , Exonucleases/genetics , Exoribonucleases , Gene Expression Profiling , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Physiologic/genetics , Oligonucleotide Array Sequence Analysis , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Up-Regulation/drug effects
20.
Blood ; 112(7): 2648-56, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18645041

ABSTRACT

Interactions between chemokines and enzymes are vital in immunoregulation. Structural protein citrullination by peptidylarginine deiminase (PAD) has been associated with autoimmunity. In this report, we identified a novel naturally occurring posttranslational modification of chemokines, that is, the deimination of arginine at position 5 into citrulline of CXC chemokine ligand 10 (CXCL10) by rabbit PAD and human PAD2. Citrullination reduced (>/= 10-fold) the chemoattracting and signaling capacity of CXCL10 for CXC chemokine receptor 3 (CXCR3) transfectants; however, it did not affect CXCR3 binding. On T lymphocytes, though, citrullinated CXCL10 remained active but was again weaker than authentic CXCL10. PAD was also able to convert CXCL11, causing an impairment of CXCR3 signaling and T-cell activation, though less pronounced than for CXCL10. Similarly, receptor binding properties of CXCL11 were not altered by citrullination. However, deimination decreased heparin binding properties of both CXCL10 and CXCL11. Overall, chemokines are the first immune modulators reported of being functionally modified by citrullination. These data provide new structure-function dimensions for chemokines in leukocyte mobilization, disclosing an anti-inflammatory role for PAD. Additionally because citrullination has severe consequences for chemokine biology, this invites to reassess the involvement and impact of PAD and citrullinated peptides in inflammation, autoimmunity, and hematologic disorders.


Subject(s)
Chemokine CXCL10/immunology , Chemokine CXCL11/immunology , Citrulline/metabolism , Hydrolases/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Cell Line , Chemokine CXCL10/chemistry , Chemokine CXCL10/isolation & purification , Chemokine CXCL10/pharmacology , Chemokine CXCL11/pharmacology , Glycosaminoglycans/metabolism , Heparin/metabolism , Humans , Lymphocyte Activation/drug effects , Molecular Sequence Data , Phytohemagglutinins/pharmacology , Protein Binding/drug effects , Protein Isoforms/metabolism , Protein Processing, Post-Translational/drug effects , Protein-Arginine Deiminase Type 2 , Protein-Arginine Deiminases , Rabbits , Receptors, CXCR/metabolism , Receptors, CXCR3/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...