Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 382
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cancer Immunol Immunother ; 72(4): 903-916, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36161509

ABSTRACT

Tumor-associated macrophages (TAMs) play an important role in tumor growth and metastasis. However, the involvement of TAMs infiltration in pulmonary osteosarcoma (OS) metastasis remains poorly understood. Therefore, the effect of OS cells on macrophages migration was investigated by in vivo and in vitro experiments to evaluate the infiltration and mechanism of TAMs in pulmonary OS metastases. The results showed that the zinc finger protein ZIM3 was upregulated in OS cells than in osteoblasts and activated the expression of CCL25, which subsequently promoted the migration of M2 macrophages. CCL25 or ZIM3 silencing in OS cells inhibited the infiltration of M2 macrophages and the formation of pulmonary metastatic nodules in a mouse model of pulmonary OS metastasis and prolonged the survival of the mice. Furthermore, bioinformatics analyses revealed that CCL25 and ZIM3 expressions are negatively correlated with the prognosis of OS patients. In conclusion, this study found that a large number of M2 TAMs were recruited into pulmonary metastatic nodules of OS through the activation of the ZIM3-CCL25 axis in OS cells, thereby facilitating OS metastasis. Therefore, the suppression of ZIM3-CCL25-induced recruitment of M2 TAMs to the metastatic sites might be considered as a therapeutic approach to inhibit the growth of pulmonary OS metastases.


Subject(s)
Bone Neoplasms , Lung Neoplasms , Osteosarcoma , Animals , Mice , Macrophages/metabolism , Cell Line, Tumor , Prognosis , Lung Neoplasms/drug therapy , Osteosarcoma/genetics , Osteosarcoma/drug therapy , Bone Neoplasms/genetics , Tumor Microenvironment , Chemokines, CC/metabolism , Chemokines, CC/pharmacology , Chemokines, CC/therapeutic use
2.
J Nanobiotechnology ; 19(1): 83, 2021 Mar 25.
Article in English | MEDLINE | ID: mdl-33766057

ABSTRACT

BACKGROUND: Chemokine therapy with C-C motif chemokine ligand 25 (CCL25) is currently under investigation as a promising approach to treat articular cartilage degeneration. We developed a delayed release mechanism based on Poly (lactic-co-glycolic acid) (PLGA) microparticle encapsulation for intraarticular injections to ensure prolonged release of therapeutic dosages. However, CCL25 plays an important role in immune cell regulation and inflammatory processes like T-cell homing and chronic tissue inflammation. Therefore, the potential of CCL25 to activate immune cells must be assessed more thoroughly before further translation into clinical practice. The aim of this study was to evaluate the reaction of different immune cell subsets upon stimulation with different dosages of CCL25 in comparison to CCL25 released from PLGA particles. RESULTS: Immune cell subsets were treated for up to 5 days with CCL25 and subsequently analyzed regarding their cytokine secretion, surface marker expression, polarization, and migratory behavior. The CCL25 receptor C-C chemokine receptor type 9 (CCR9) was expressed to a different extent on all immune cell subsets. Direct stimulation of peripheral blood mononuclear cells (PBMCs) with high dosages of CCL25 resulted in strong increases in the secretion of monocyte chemoattractant protein-1 (MCP-1), interleukin-8 (IL-8), interleukin-1ß (IL-1ß), tumor-necrosis-factor-α (TNF-α) and interferon-γ (IFN-γ), upregulation of human leukocyte antigen-DR (HLA-DR) on monocytes and CD4+ T-cells, as well as immune cell migration along a CCL25 gradient. Immune cell stimulation with the supernatants from CCL25 loaded PLGA microparticles caused moderate increases in MCP-1, IL-8, and IL-1ß levels, but no changes in surface marker expression or migration. Both CCL25-loaded and unloaded PLGA microparticles induced an increase in IL-8 and MCP-1 release in PBMCs and macrophages, and a slight shift of the surface marker profile towards the direction of M2-macrophage polarization. CONCLUSIONS: While supernatants of CCL25 loaded PLGA microparticles did not provoke strong inflammatory reactions, direct stimulation with CCL25 shows the critical potential to induce global inflammatory activation of human leukocytes at certain concentrations. These findings underline the importance of a safe and reliable release system in a therapeutic setup. Failure of the delivery system could result in strong local and systemic inflammatory reactions that could potentially negate the benefits of chemokine therapy.


Subject(s)
Chemokines, CC/pharmacology , Chemokines, CC/therapeutic use , Delayed-Action Preparations/pharmacology , Delayed-Action Preparations/therapeutic use , Inflammation/drug therapy , Chemokine CCL2/metabolism , Chemokines/pharmacology , Chemokines/therapeutic use , Humans , Interferon-gamma , Interleukin-1beta/metabolism , Interleukin-8/metabolism , Leukocytes, Mononuclear , Ligands , Macrophages/metabolism , Monocytes , Polylactic Acid-Polyglycolic Acid Copolymer , Receptors, CCR/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
Article in English | MEDLINE | ID: mdl-32423961

ABSTRACT

Candida albicans is a commensal organism that causes life-threatening or life-altering opportunistic infections. Treatment of Candida infections is limited by the paucity of antifungal drug classes. Naturally occurring antimicrobial peptides are promising agents for drug development. CCL28 is a CC chemokine that is abundant in saliva and has in vitro antimicrobial activity. In this study, we examine the in vivo Candida killing capacity of CCL28 in oropharyngeal candidiasis as well as the spectrum and mechanism of anti-Candida activity. In the mouse model of oropharyngeal candidiasis, application of wild-type CCL28 reduces oral fungal burden in severely immunodeficient mice without causing excessive inflammation or altering tissue neutrophil recruitment. CCL28 is effective against multiple clinical strains of C. albicans Polyamine protein transporters are not required for CCL28 anti-Candida activity. Both structured and unstructured CCL28 proteins show rapid and sustained fungicidal activity that is superior to that of clinical antifungal agents. Application of wild-type CCL28 to C. albicans results in membrane disruption as measured by solute movement, enzyme leakage, and induction of negative Gaussian curvature on model membranes. Membrane disruption is reduced in CCL28 lacking the functional C-terminal tail. Our results strongly suggest that CCL28 can exert antifungal activity in part via membrane permeation and has potential for development as an anti-Candida therapeutic agent without inflammatory side effects.


Subject(s)
Antifungal Agents , Candidiasis, Oral , Chemokines, CC/pharmacology , Animals , Antifungal Agents/pharmacology , Candida albicans , Candidiasis, Oral/drug therapy , Chemokines , Mice , Microbial Sensitivity Tests
4.
Stem Cells ; 37(3): 407-416, 2019 03.
Article in English | MEDLINE | ID: mdl-30485583

ABSTRACT

Understanding the mechanisms regulating recruitment of human skeletal (stromal or mesenchymal) stem cells (hMSC) to sites of tissue injury is a prerequisite for their successful use in cell replacement therapy. Chemokine-like protein TAFA2 is a recently discovered neurokine involved in neuronal cell migration and neurite outgrowth. Here, we demonstrate a possible role for TAFA2 in regulating recruitment of hMSC to bone fracture sites. TAFA2 increased the in vitro trans-well migration and motility of hMSC in a dose-dependent fashion and induced significant morphological changes including formation of lamellipodia as revealed by high-content-image analysis at single-cell level. Mechanistic studies revealed that TAFA2 enhanced hMSC migration through activation of the Rac1-p38 pathway. In addition, TAFA2 enhanced hMSC proliferation, whereas differentiation of hMSC toward osteoblast and adipocyte lineages was not altered. in vivo studies demonstrated transient upregulation of TAFA2 gene expression during the inflammatory phase of fracture healing in a closed femoral fracture model in mice, and a similar pattern was observed in serum levels of TAFA2 in patients after hip fracture. Finally, interleukin-1ß was found as an upstream regulator of TAFA2 expression. Our findings demonstrate that TAFA2 enhances hMSC migration and recruitment and thus is relevant for regenerative medicine applications. Stem Cells 2019;37:407-416.


Subject(s)
Cell Movement/drug effects , Chemokines, CC/pharmacology , MAP Kinase Signaling System/drug effects , Mesenchymal Stem Cells/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chemokines, CC/metabolism , Disease Models, Animal , Hip Fractures/metabolism , Hip Fractures/pathology , Humans , Mesenchymal Stem Cells/pathology , Mice , Neuropeptides/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology
5.
J Cell Physiol ; 232(11): 2996-3005, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28387445

ABSTRACT

The focus of this study was to determine which chemokine receptors are present on oral fibroblasts and whether these receptors influence proliferation, migration, and/or the release of wound healing mediators. This information may provide insight into the superior wound healing characteristics of the oral mucosa. The gingiva fibroblasts expressed 12 different chemokine receptors (CCR3, CCR4, CCR6, CCR9, CCR10, CXCR1, CXCR2, CXCR4, CXCR5, CXCR7, CX3CR1, and XCR1), as analyzed by flow cytometry. Fourteen corresponding chemokines (CCL5, CCL15, CCL20, CCL22, CCL25, CCL27, CCL28, CXCL1, CXCL8, CXCL11, CXCL12, CXCL13, CX3CL1, and XCL1) were used to study the activation of these receptors on gingiva fibroblasts. Twelve of these fourteen chemokines stimulated gingiva fibroblast migration (all except for CXCL8 and CXCL12). Five of the chemokines stimulated proliferation (CCL5/CCR3, CCL15/CCR3, CCL22/CCR4, CCL28/CCR3/CCR10, and XCL1/XCR1). Furthermore, CCL28/CCR3/CCR10 and CCL22/CCR4 stimulation increased IL-6 secretion and CCL28/CCR3/CCR10 together with CCL27/CCR10 upregulated HGF secretion. Moreover, TIMP-1 secretion was reduced by CCL15/CCR3. In conclusion, this in-vitro study identifies chemokine receptor-ligand pairs which may be used in future targeted wound healing strategies. In particular, we identified the chemokine receptors CCR3 and CCR4, and the mucosa specific chemokine CCL28, as having an predominant role in oral wound healing by increasing human gingiva fibroblast proliferation, migration, and the secretion of IL-6 and HGF and reducing the secretion of TIMP-1.


Subject(s)
Chemokines, CC/pharmacology , Fibroblasts/drug effects , Gingiva/drug effects , Receptors, CCR3/agonists , Receptors, CCR4/agonists , Wound Healing/drug effects , Cell Line, Transformed , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Fibroblasts/pathology , Gingiva/metabolism , Gingiva/pathology , Hepatocyte Growth Factor/metabolism , Humans , Interleukin-6/metabolism , Ligands , Receptors, CCR3/metabolism , Receptors, CCR4/metabolism , Signal Transduction/drug effects , Tissue Inhibitor of Metalloproteinase-1/metabolism
6.
Int J Mol Sci ; 18(7)2017 Jul 20.
Article in English | MEDLINE | ID: mdl-28726743

ABSTRACT

Despite effective treatment for those living with Human Immunodeficiency Virus (HIV), there are still two million new infections each year. Protein-based HIV entry inhibitors, being highly effective and specific, could be used to protect people from initial infection. One of the most promising of these for clinical use is 5P12-RANTES, a variant of the chemokine RANTES/CCL5. The N-terminal amino acid of 5P12-RANTES is glutamine (Gln; called Q0), a residue that is prone to spontaneous cyclization when at the N-terminus of a protein. It is not known how this cyclization affects the potency of the inhibitor or whether cyclization is necessary for the function of the protein, although the N-terminal region of RANTES has been shown to be critical for receptor interactions, with even small changes having a large effect. We have studied the kinetics of cyclization of 5P12-RANTES as well as N-terminal variations of the protein that either produce an identical cyclized terminus (Glu0) or that cannot similarly cyclize (Asn0, Phe0, Ile0, and Leu0). We find that the half life for N-terminal cyclization of Gln is roughly 20 h at pH 7.3 at 37 °C. However, our results show that cyclization is not necessary for the potency of this protein and that several replacement terminal amino acids produce nearly-equally potent HIV inhibitors while remaining CC chemokine receptor 5 (CCR5) antagonists. This work has ramifications for the production of active 5P12-RANTES for use in the clinic, while also opening the possibility of developing other inhibitors by varying the N-terminus of the protein.


Subject(s)
Chemokines, CC/chemistry , Chemokines, CC/pharmacology , HIV Fusion Inhibitors/chemistry , HIV Fusion Inhibitors/pharmacology , Amino Acid Motifs , Animals , CHO Cells , Cell Line , Cricetulus , Cyclization , HIV-1/drug effects , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Molecular Structure , Structure-Activity Relationship , Virus Internalization/drug effects
7.
Protein Expr Purif ; 119: 1-10, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26506568

ABSTRACT

In the continued absence of an effective anti-HIV vaccine, approximately 2 million new HIV infections occur every year, with over 95% of these in developing countries. Calls have been made for the development of anti-HIV drugs that can be formulated for topical use to prevent HIV transmission during sexual intercourse. Because these drugs are principally destined for use in low-resource regions, achieving production costs that are as low as possible is an absolute requirement. 5P12-RANTES, an analog of the human chemokine protein RANTES/CCL5, is a highly potent HIV entry inhibitor which acts by achieving potent blockade of the principal HIV coreceptor, CCR5. Here we describe the development and optimization of a scalable low-cost production process for 5P12-RANTES based on expression in Pichia pastoris. At pilot (150 L) scale, this cGMP compliant process yielded 30 g of clinical grade 5P12-RANTES. As well as providing sufficient material for the first stage of clinical development, this process represents an important step towards achieving production of 5P12-RANTES at a cost and scale appropriate to meet needs for topical HIV prevention worldwide.


Subject(s)
Anti-HIV Agents/metabolism , Chemokines, CC/biosynthesis , HIV Infections/drug therapy , HIV/drug effects , Pichia , Anti-HIV Agents/isolation & purification , Anti-HIV Agents/pharmacology , Bioreactors/economics , Bioreactors/standards , Chemokines, CC/isolation & purification , Chemokines, CC/pharmacology , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Fermentation , Humans , Inhibitory Concentration 50 , Pilot Projects , Virus Internalization/drug effects
8.
Blood ; 121(19): 3838-42, S1-15, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23509159

ABSTRACT

In an attempt to discover novel growth factors for hematopoietic stem and progenitor cells (HSPCs), we have assessed cytokine responses of cord blood (CB)-derived CD34(+) cells in a high-content growth factor screen. We identify the immunoregulatory chemokine (C-C motif) ligand 28 (CCL28) as a novel growth factor that directly stimulates proliferation of primitive hematopoietic cells from different ontogenetic origins. CCL28 enhances the functional progenitor cell content of cultured cells by stimulating cell cycling and induces gene expression changes associated with survival. Importantly, addition of CCL28 to cultures of purified putative hematopoietic stem cells (HSCs) significantly increases the ability of the cells to long-term repopulate immunodeficient mice compared with equivalent input numbers of fresh cells. Together, our findings identify CCL28 as a potent growth-promoting factor with the ability to support the in vitro and in vivo functional properties of cultured human hematopoietic cells.


Subject(s)
Cell Proliferation , Chemokines, CC/physiology , Hematopoietic Stem Cells/physiology , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Chemokines, CC/genetics , Chemokines, CC/metabolism , Chemokines, CC/pharmacology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Infant, Newborn , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/physiology , Mice , Mice, Inbred NOD , Mice, Transgenic , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/physiology
9.
Allergy ; 70(2): 161-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25377782

ABSTRACT

BACKGROUND: During bacterial infections of the airways, a Th1-profiled inflammation promotes the production of several host defense proteins and peptides with antibacterial activities including ß-defensins, ELR-negative CXC chemokines, and the cathelicidin LL-37. These are downregulated by Th2 cytokines of the allergic response. Instead, the eosinophil-recruiting chemokines eotaxin-1/CCL11, eotaxin-2/CCL24, and eotaxin-3/CCL26 are expressed. This study set out to investigate whether these chemokines could serve as innate host defense molecules during allergic inflammation. METHODS: Antibacterial activities of the eotaxins were investigated using viable count assays, electron microscopy, and methods assessing bacterial permeabilization. Fragments generated by mast cell proteases were characterized, and their potential antibacterial, receptor-activating, and lipopolysaccharide-neutralizing activities were investigated. RESULTS: CCL11, CCL24, and CCL26 all showed potent bactericidal activity, mediated through membrane disruption, against the airway pathogens Streptococcus pneumoniae, Staphylococcus aureus, Nontypeable Haemophilus influenzae, and Pseudomonas aeruginosa. CCL26 retained bactericidal activity in the presence of salt at physiologic concentrations, and the region holding the highest bactericidal activity was the cationic and amphipathic COOH-terminus. Proteolysis of CCL26 by chymase and tryptase, respectively, released distinct fragments of the COOH- and NH2 -terminal regions. The COOH-terminal fragment retained antibacterial activity while the NH2 -terminal had potent LPS-neutralizing properties in the order of CCL26 full-length protein. An identical fragment to NH2 -terminal fragment generated by tryptase was obtained after incubation with supernatants from activated mast cells. None of the fragments activated the CCR3-receptor. CONCLUSIONS: Taken together, the findings show that the eotaxins can contribute to host defense against common airway pathogens and that their activities are modulated by mast cell proteases.


Subject(s)
Chemokines, CC/metabolism , Immunity, Innate , Mast Cells/immunology , Mast Cells/metabolism , Peptide Hydrolases/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Cell Membrane/drug effects , Chemokine CCL11/metabolism , Chemokine CCL11/pharmacology , Chemokine CCL24/metabolism , Chemokine CCL24/pharmacology , Chemokine CCL26 , Chemokines, CC/chemistry , Chemokines, CC/pharmacology , Humans , Models, Molecular , Peptide Hydrolases/chemistry , Protein Conformation , Receptors, CCR3/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/ultrastructure , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/ultrastructure
10.
Protein Expr Purif ; 113: 1-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25979463

ABSTRACT

FAM19A4 is a novel potential cytokine identified by our group, which can chemoattract macrophages, promote phagocytosis against zymosan and increase reactive oxygen species (ROS) release. To further explore the role of FAM19A4 in immune system, abundant recombinant protein with high quality is indispensable. For efficient production of FAM19A4, we used an improved CHO-S cell expression system on the basis of pMH3 vector containing GC-rich regions which were novel ubiquitous chromatin opening elements (UCOEs). We selected CHO-S cells stably expressing FAM19A4 with G418 and screened cell clones with high level of FAM19A4 expression by immune blot and his-ELISA, adapted cell clones to serum-free suspension culture. Afterwards, we obtained the highest FAM19A4 expressing cell clone (2#) through 40 ml batch culture. We optimized the fed-batch culture condition and discovered the final cell viability was critical for FAM19A4 production successfully. Then we scaled 2# clone up to 3 L in fed-batch culture and obtained 22 mg (7.33 mg/L, averagely) endotoxin free FAM19A4 protein with purity over 95% using Ni affinity chromatography and size exclusion chromatography. The final yield was increased 3.6-folds compared to that of our previously reported transient system. Besides, the purified FAM19A4 protein showed chemotactic activity on macrophages. In summary, we developed a stable optimized fed-batch CHO-S cell system to produce FAM19A4, which not only provided sufficient bioactive FAM19A4 protein for further research but also offered an efficient strategy for other recombinant protein production.


Subject(s)
Chemokines, CC/metabolism , Recombinant Proteins/metabolism , Animals , Bioreactors , CHO Cells , Chemokines, CC/chemistry , Chemokines, CC/genetics , Chemokines, CC/isolation & purification , Chemokines, CC/pharmacology , Chemotaxis/drug effects , Chromatography, Liquid , Cricetinae , Cricetulus , Genetic Vectors , Humans , Macrophages, Peritoneal , Monocytes , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
11.
J Biol Chem ; 288(10): 7169-81, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23341447

ABSTRACT

Chemokine receptors form a large subfamily of G protein-coupled receptors that predominantly activate heterotrimeric Gi proteins and are involved in immune cell migration. CCX-CKR is an atypical chemokine receptor with high affinity for CCL19, CCL21, and CCL25 chemokines, but is not known to activate intracellular signaling pathways. However, CCX-CKR acts as decoy receptor and efficiently internalizes these chemokines, thereby preventing their interaction with other chemokine receptors, like CCR7 and CCR9. Internalization of fluorescently labeled CCL19 correlated with ß-arrestin2-GFP translocation. Moreover, recruitment of ß-arrestins to CCX-CKR in response to CCL19, CCL21, and CCL25 was demonstrated using enzyme-fragment complementation and bioluminescence resonance energy transfer methods. To unravel why CCX-CKR is unable to activate Gi signaling, CCX-CKR chimeras were constructed by substituting its intracellular loops with the corresponding CCR7 or CCR9 domains. The signaling properties of chimeric CCX-CKR receptors were characterized using a cAMP-responsive element (CRE)-driven reporter gene assay. Unexpectedly, wild type CCX-CKR and a subset of the chimeras induced an increase in CRE activity in response to CCL19, CCL21, and CCL25 in the presence of the Gi inhibitor pertussis toxin. CCX-CKR signaling to CRE required an intact DRY motif. These data suggest that inactive Gi proteins impair CCX-CKR signaling most likely by hindering the interaction of this receptor with pertussis toxin-insensitive G proteins that transduce signaling to CRE. On the other hand, recruitment of the putative signaling scaffold ß-arrestin to CCX-CKR in response to chemokines might allow activation of yet to be identified signal transduction pathways.


Subject(s)
Arrestins/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptors, CCR/metabolism , Signal Transduction , Animals , Arrestins/genetics , Binding, Competitive/drug effects , Blotting, Western , CHO Cells , Cell Line, Tumor , Chemokine CCL19/metabolism , Chemokine CCL19/pharmacology , Chemokine CCL21/metabolism , Chemokine CCL21/pharmacology , Chemokines, CC/metabolism , Chemokines, CC/pharmacology , Cricetinae , Cricetulus , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Microscopy, Fluorescence , Models, Biological , Pertussis Toxin/pharmacology , Protein Binding/drug effects , Protein Transport/drug effects , Receptors, CCR/genetics , beta-Arrestins
12.
J Virol ; 87(14): 7911-20, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23658453

ABSTRACT

Murine cytomegalovirus encodes numerous proteins that act on a variety of pathways to modulate the innate and adaptive immune responses. Here, we demonstrate that a chemokine-like protein encoded by murine cytomegalovirus activates the early innate immune response and delays adaptive immunity, thereby impairing viral clearance. The protein, m131/129 (also known as MCK-2), is not required to establish infection in the spleen; however, a mutant virus lacking m131/129 was cleared more rapidly from this organ. In the absence of m131/129 expression, there was enhanced activation of dendritic cells (DC), and virus-specific CD8(+) T cells were recruited into the immune response earlier. Viral mutants lacking m131/129 elicited weaker production of alpha interferon (IFN-α) at 40 h postinfection, indicating that this protein exerts its effects during early rounds of viral replication in the spleen. Furthermore, while wild-type and mutant viruses activated plasmacytoid dendritic cells (pDC) equally at this time, as measured by the upregulation of costimulatory molecules, the presence of m131/129 stimulated more pDC to secrete IFN-α, accounting for the stronger IFN-α response than from the wild-type virus. These data provide evidence for a novel immunomodulatory function of a viral chemokine and expose the multifunctionality of immune evasion proteins. In addition, these results broaden our understanding of the interplay between innate and adaptive immunity.


Subject(s)
Adaptive Immunity/drug effects , Chemokines, CC/pharmacology , Dendritic Cells/drug effects , Immune Evasion/immunology , Immunity, Innate/drug effects , Muromegalovirus/immunology , Viral Proteins/pharmacology , Animals , CD8-Positive T-Lymphocytes/immunology , Chemokines/immunology , Chemokines, CC/immunology , Cytokines/immunology , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Interferon-alpha/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Spleen/immunology , Statistics, Nonparametric , Viral Proteins/immunology
13.
Stem Cells ; 31(9): 1980-91, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23712682

ABSTRACT

Multipotent stromal cells (MSC) have been shown to possess immunomodulatory capacities and are therefore explored as a novel cellular therapy. One of the mechanisms through which MSC modulate immune responses is by the promotion of regulatory T cell (Treg) formation. In this study, we focused on the cellular interactions and secreted factors that are essential in this process. Using an in vitro culture system, we showed that culture-expanded bone marrow-derived MSC promote the generation of CD4(+) CD25(hi) FoxP3(+) T cells in human PBMC populations and that these populations are functionally suppressive. Similar results were obtained with MSC-conditioned medium, indicating that this process is dependent on soluble factors secreted by the MSC. Antibody neutralization studies showed that TGF-ß1 mediates induction of Tregs. TGF-ß1 is constitutively secreted by MSC, suggesting that the MSC-induced generation of Tregs by TGF-ß1 was independent of the interaction between MSC and PBMC. Monocyte-depletion studies showed that monocytes are indispensable for MSC-induced Treg formation. MSC promote the survival of monocytes and induce differentiation toward macrophage type 2 cells that express CD206 and CD163 and secrete high levels of IL-10 and CCL-18, which is mediated by as yet unidentified MSC-derived soluble factors. CCL18 proved to be responsible for the observed Treg induction. These data indicate that MSC promote the generation of Tregs. Both the direct pathway through the constitutive production of TGF-ß1 and the indirect novel pathway involving the differentiation of monocytes toward CCL18 producing type 2 macrophages are essential for the generation of Tregs induced by MSC.


Subject(s)
Inflammation/pathology , Macrophages/pathology , Monocytes/cytology , Multipotent Stem Cells/cytology , T-Lymphocytes, Regulatory/cytology , CD4 Antigens/metabolism , Cell Differentiation/drug effects , Cell Survival/drug effects , Chemokines, CC/pharmacology , Coculture Techniques , Culture Media, Conditioned/pharmacology , Forkhead Transcription Factors/metabolism , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Macrophages/drug effects , Macrophages/metabolism , Monocytes/drug effects , Monocytes/metabolism , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Signal Transduction/drug effects , Solubility , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta1/metabolism
14.
J Immunol ; 189(11): 5266-76, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23125416

ABSTRACT

Agonists of CCR1 contribute to hypersensitivity reactions and atherosclerotic lesions, possibly via the regulation of the transcription factor STAT3. CCR1 was demonstrated to use pertussis toxin-insensitive Gα(14/16) to stimulate phospholipase Cß and NF-κB, whereas both Gα(14) and Gα(16) are also capable of activating STAT3. The coexpression of CCR1 and Gα(14/16) in human THP-1 macrophage-like cells suggests that CCR1 may use Gα(14/16) to induce STAT3 activation. In this study, we demonstrated that a CCR1 agonist, leukotactin-1 (CCL15), could indeed stimulate STAT3 Tyr(705) and Ser(727) phosphorylation via pertussis toxin-insensitive G proteins in PMA-differentiated THP-1 cells, human erythroleukemia cells, and HEK293 cells overexpressing CCR1 and Gα(14/16). The STAT3 Tyr(705) and Ser(727) phosphorylations were independent of each other and temporally distinct. Subcellular fractionation and confocal microscopy illustrated that Tyr(705)-phosphorylated STAT3 translocated to the nucleus, whereas Ser(727)-phosphorylated STAT3 was retained in the cytosol after CCR1/Gα(14) activation. CCL15 was capable of inducing IL-6 and IL-8 (CXCL8) production in both THP-1 macrophage-like cells and HEK293 cells overexpressing CCR1 and Gα(14/16). Neutralizing Ab to IL-6 inhibited CCL15-mediated STAT3 Tyr(705) phosphorylation, whereas inhibition of STAT3 activity abolished CCL15-activated CXCL8 release. The ability of CCR1 to signal through Gα(14/16) provides a linkage for CCL15 to regulate IL-6/STAT3-signaling cascades, leading to expression of CXCL8, a cytokine that is involved in inflammation and the rupture of atherosclerotic plaque.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/immunology , Interleukin-6/immunology , Interleukin-8/immunology , Macrophages/immunology , Receptors, CCR1/immunology , STAT3 Transcription Factor/immunology , Antibodies/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/immunology , Chemokines, CC/immunology , Chemokines, CC/pharmacology , Cytosol/drug effects , Cytosol/immunology , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Gene Expression/drug effects , Gene Expression/immunology , HEK293 Cells , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-8/biosynthesis , K562 Cells , Macrophage Inflammatory Proteins/immunology , Macrophage Inflammatory Proteins/pharmacology , Macrophages/cytology , Macrophages/drug effects , Pertussis Toxin/pharmacology , Phosphorylation , Plasmids , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptors, CCR1/agonists , Receptors, CCR1/genetics , STAT3 Transcription Factor/genetics , Serine/metabolism , Signal Transduction/drug effects , Transfection , Tyrosine/metabolism
15.
J Struct Biol ; 184(2): 329-34, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24091038

ABSTRACT

Clustering of arrestins upon G protein-coupled receptor stimulation is a phenomenon that is well-known but difficult to describe quantitatively due to the size of the clusters close to the diffraction limit of visible light. We introduce a general method to quantitatively investigate the clustering of arrestin following stimulation of the C-C chemokine receptor 5 (CCR5) using single-molecule super-resolution imaging and coordinate and image-based cluster analysis. We investigated the effect of potent anti-HIV ligands of CCR5 with different pharmacological profiles on arrestin2 cluster formation and found that only the ligands capable of inducing CCR5 internalization induced arrestin2 recruitment and clustering. We further demonstrate that the fraction of arrestin2 molecules found in clusters larger than 100nm correlates with the magnitude of ligand-induced CCR5 internalization, but not with G protein activation, indicating that recruitment of arrestin2 to CCR5 is independent of G protein activation. Pre-treatment of the cells with the drug cytochalasin D, which blocks actin polymerization, led to the formation of larger clusters, whereas the inhibitor of microtubule polymerization nocodazole had little effect on arrestin2 recruitment, suggesting an active role of actin in the organization and dynamics of these aggregates.


Subject(s)
Arrestins/metabolism , Chemokine CCL5/physiology , Receptors, CCR5/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , CHO Cells , Cattle , Chemokine CCL5/pharmacology , Chemokines, CC/pharmacology , Cricetinae , Cricetulus , Cytochalasin D/pharmacology , Green Fluorescent Proteins/metabolism , Microscopy, Fluorescence , Nocodazole/pharmacology , Protein Transport , Recombinant Fusion Proteins/metabolism , Single-Domain Antibodies/chemistry , Tubulin Modulators/pharmacology
16.
Blood ; 118(13): 3549-58, 2011 Sep 29.
Article in English | MEDLINE | ID: mdl-21803856

ABSTRACT

The aim of this study was to evaluate the nonchemotactic function of CCL18 on human dendritic cells (DCs). In different protocols of DC differentiation, CCL18 was highly produced, suggesting that it may constitute a mandatory mediator of the differentiation process. Differentiation of monocytes from healthy subjects in the presence of granulocyte-macrophage colony-stimulating factor and CCL18 led to the development of DCs with a semimature phenotype, with intermediate levels of costimulatory and MHC class II molecules, increased CCR7 expression, which induced, in coculture with allogenic naive T cells, an increase in IL-10 production. The generated T cells were able to suppress the proliferation of effector CD4(+)CD25(-) cells, through a cytokine-dependent mechanism, and exhibited characteristics of type 1 T regulatory cells. The generation of tolerogenic DCs by CCL18 was dependent on the production of indoleamine 2,3-dioxigenase through an interleukin-10-mediated mechanism. Surprisingly, when DCs originated from allergic patients, the tolerogenic effect of CCL18 was lost in relation with a decreased binding of CCL18 to its putative receptor. This study is the first to define a chemokine able to generate tolerogenic DCs. However, this function was absent in allergic donors and may participate to the decreased tolerance observed in allergic diseases.


Subject(s)
Antigen Presentation/drug effects , Cell Differentiation/drug effects , Chemokines, CC/pharmacology , Dendritic Cells/drug effects , Immune Tolerance/drug effects , T-Lymphocytes, Regulatory/drug effects , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/physiology , Cell Differentiation/genetics , Cells, Cultured , Chemokines, CC/genetics , Chemokines, CC/metabolism , Chemokines, CC/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/physiology , Forkhead Transcription Factors/metabolism , Health , Humans , Hypersensitivity/immunology , Hypersensitivity/metabolism , Immune Tolerance/genetics , Immune Tolerance/immunology , Interleukin-10/metabolism , Primary Cell Culture , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
17.
Hum Reprod ; 28(6): 1497-507, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23477905

ABSTRACT

STUDY QUESTION: What are the effects of the eotaxin group of chemokines (CCL11, CCL24 and CCL26) on extravillous trophoblast (EVT) functions important during uterine decidual vessel remodelling? SUMMARY ANSWER: CCL11, CCL24 and CCL26 can regulate EVT migration, invasion and adhesion, highlighting a potential regulatory role for these chemokines during uterine decidual spiral arteriole remodelling in the first trimester of human pregnancy. WHAT IS KNOWN ALREADY: A successful human pregnancy depends on adequate remodelling of the uterine decidual spiral arterioles, a process carried out by EVT which invade from the placenta. The invasion by EVT into the maternal uterine decidual vessels is regulated by the interaction of many factors including members of the chemokine subfamily of cytokines. STUDY DESIGN, SIZE, DURATION: This study used the HTR8/SVneo cell line as a model for invasive EVT. All experiments were repeated on at least three separate occasions. PARTICIPANTS/MATERIALS, SETTING, METHODS: The effect of recombinant human CCL11, CCL24 and CCL26 on EVT migration and invasive potential was measured using the xCELLigence real-time system, wound-healing and Matrigel invasion assays, zymography to measure MMP activity and reverse zymography to measure TIMP activity. A commercially available adhesion assay was used to assess EVT adhesion to extracellular matrix proteins. MAIN RESULTS AND THE ROLE OF CHANCE: All the three eotaxins were found to significantly stimulate migration of the EVT-derived cell line HTR8/SVneo (P < 0.05) with no significant changes in cell number following treatment with each chemokine (P > 0.05). All the three eotaxins significantly increased HTR8/SVneo invasion (P < 0.05) and MMP2 activity (P < 0.05) without any effects on TIMP2 activity (P > 0.05). All the three eotaxins significantly increased HTR8/SVneo cell binding to collagen IV (P < 0.05) and fibronectin (P < 0.05). LIMITATIONS, REASONS FOR CAUTION: This work has been conducted in vitro with a commonly used cell line model of EVT, HTR8/SVneo. WIDER IMPLICATIONS OF THE FINDINGS: This study is the first to comprehensively examine the effects of the eotaxin group of chemokines on EVT functions and demonstrates that all the three eotaxins have the ability to regulate EVT functions critical to their role in vessel remodelling. This identifies a new role for the eotaxin group of chemokines during placentation.


Subject(s)
Chemokine CCL11/pharmacology , Chemokine CCL24/pharmacology , Chemokines, CC/pharmacology , Decidua/drug effects , Trophoblasts/drug effects , Arterioles/drug effects , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Chemokine CCL11/physiology , Chemokine CCL24/physiology , Chemokine CCL26 , Chemokines, CC/physiology , Collagen , Decidua/blood supply , Drug Combinations , Female , Humans , Laminin , Proteoglycans , Trophoblasts/cytology
18.
Nat Commun ; 14(1): 7940, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38040762

ABSTRACT

The C-C motif chemokine receptor 8 (CCR8) is a class A G-protein coupled receptor that has emerged as a promising therapeutic target in cancer. Targeting CCR8 with an antibody has appeared to be an attractive therapeutic approach, but the molecular basis for chemokine-mediated activation and antibody-mediated inhibition of CCR8 are not fully elucidated. Here, we obtain an antagonist antibody against human CCR8 and determine structures of CCR8 in complex with either the antibody or the endogenous agonist ligand CCL1. Our studies reveal characteristic antibody features allowing recognition of the CCR8 extracellular loops and CCL1-CCR8 interaction modes that are distinct from other chemokine receptor - ligand pairs. Informed by these structural insights, we demonstrate that CCL1 follows a two-step, two-site binding sequence to CCR8 and that antibody-mediated inhibition of CCL1 signaling can occur by preventing the second binding event. Together, our results provide a detailed structural and mechanistic framework of CCR8 activation and inhibition that expands our molecular understanding of chemokine - receptor interactions and offers insight into the development of therapeutic antibodies targeting chemokine GPCRs.


Subject(s)
Chemokines, CC , Receptors, Chemokine , Humans , Chemokines, CC/metabolism , Chemokines, CC/pharmacology , Receptors, CCR8/genetics , Ligands , Chemokine CCL1/metabolism , Receptors, Chemokine/genetics , Antibodies
19.
Radiat Res ; 200(3): 281-288, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37450610

ABSTRACT

Connexin26 (Cx26) plays an important role in ionizing radiation-induced damage, and CC chemokine ligand 27 (CCL27) regulates the skin immune response. However, the relationship between Cx26 and CCL27 in radiation-induced skin damage is unclear. After X-ray irradiation, clonogenic survival and micronucleus formation were assessed in immortalized human keratinocytes (HaCaT). Proteins in the mitogen activated protein kinase (MAPK) signaling pathway and CCL27-related proteins were detected by immunoblotting. HaCaTCx26-/- cells were constructed to verify the effects of Cx26 on CCL27 secretion. A mouse model was established to examine the expression of CCL27 and skin inflammation in vivo. The degree of skin injury induced by 6 MV of X rays was closely related to CCL27. The phosphorylation of ERK, p38 and NF-κB was significantly increased in irradiated cells. The secretion of CCL27 was significantly decreased in HaCaT wild-type cells relative to HaCaTCx26-/- cells. Whereas cell survival fractions decreased, and the micronuclei formation rate increased as a function of increasing X-ray dose in HaCaT cells, the opposite trend occurred in HaCaTCx26-/- cells. Our findings show that Cx26 likely plays a role in the activation of the MAPK and NF-κB/COX-2 signaling pathways and regulates the secretion of CCL27 in keratinocytes after X-ray radiation-induced skin damage.


Subject(s)
Chemokine CCL27 , Radiodermatitis , Animals , Humans , Mice , Chemokine CCL27/metabolism , Chemokine CCL27/pharmacology , Chemokines/metabolism , Chemokines, CC/metabolism , Chemokines, CC/pharmacology , Keratinocytes/metabolism , Ligands , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/pharmacology , NF-kappa B/metabolism , Radiodermatitis/etiology , Signal Transduction
20.
Immunology ; 135(4): 287-98, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22117697

ABSTRACT

The observation that human monocytes cultured in the presence of the chemokine CCL18 showed increased survival, led us to profile cytokine expression in CCL18-stimulated versus control cultures. CCL18 caused significantly increased expression of chemokines (CXCL8, CCL2, CCL3 and CCL22), interleukin-10 (IL-10) and platelet-derived growth factor, but no up-regulation of M1 cytokines IL-1ß or IL-12. CCL18-stimulated monocytes matured into cells with morphological resemblance to IL-4-stimulated macrophages, and expressed the monocyte marker CD14 as well the M2 macrophage markers CD206 and 15-lipoxygenase, but no mature dendritic cell markers (CD80, CD83 or CD86). Functionally, CCL18-stimulated macrophages showed a high capacity for unspecific phagocytosis and for pinocytosis, which was not associated with an oxidative burst. These findings suggest that CCL18-activated macrophages stand at the cross-roads between inflammation and its resolution. The chemokines that are produced in response to CCL18 are angiogenic and attract various leucocyte populations, which sustain inflammation. However, the capacity of these cells to remove cellular debris without causing oxidative damage and the production of the anti-inflammatory IL-10 will initiate termination of the inflammatory response. In summary, CCL18 induces an M2 spectrum macrophage phenotype in the absence of IL-4.


Subject(s)
Cell Differentiation/drug effects , Chemokines, CC/pharmacology , Macrophages/immunology , Monocytes/cytology , Animals , Cells, Cultured , Chemokines/metabolism , Chemokines, CC/immunology , Guinea Pigs , Humans , Interleukin-10/metabolism , Macrophages/cytology , Mice , Monocytes/drug effects , Monocytes/immunology , Phagocytosis
SELECTION OF CITATIONS
SEARCH DETAIL