Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 367: 114451, 2023 09.
Article in English | MEDLINE | ID: mdl-37257716

ABSTRACT

AIMS: Intracerebral hemorrhage (ICH) is a severe neurological condition with high mortality and morbidity. Microglia activation and peripheral inflammatory cells infiltration play an important role in ICH prognosis. Previous studies demonstrated that regulatory T cells (Tregs) ameliorated neuroinflammation following experimental ICH. However, the molecular mechanism underlying such effects of Tregs remains unclear. The objective was to examine how Tregs recruitment induced by recombinant CC chemokine ligand 17 (rCCL17) influences microglia/macrophage polarization in an intrastriatal autologous blood injection ICH animal model, and to determine if TGFß/TGFß-R/Smad2/3 pathway was involved. METHODS: 380 adult CD1 mice (male, eight weeks old) were subjected to sham surgery or autologous blood injection induced ICH. A CD25-specific mouse antibody or isotype control mAb was injected intraventricular (i.c.v) 48 h prior to ICH induction to deplete Tregs. rCCL17, a CC chemokine receptor 4 (CCR4) ligand, was delivered intranasally at 1 h post-ICH. SB431542, a specific inhibitor of TGF-ß was administered intraperitoneally 1 h before ICH induction. Following the ICH, neurobehavioral testing, brain edema, hematoma volume, hemoglobin content, western blotting, double immunofluorescence labeling, and immunohistochemistry were performed. RESULTS: Endogenous expressions of CCL17, Tregs marker Foxp3, and the number of Tregs in perihematomal region increased following ICH. Tregs depletion with a CD25 antibody aggravated neurological deficits and brain edema, increased inflammatory cytokines, neutrophil infiltration, oxidative stress, and reduced the rate of hematoma resolution in ICH mice. rCCL17 treatment increased the number of Tregs in the brain, ameliorated neurological deficits and brain edema after ICH, and promoted microglia/macrophage polarization toward M2 phenotype which was reversed with CD25 antibody. Moreover, rCCL17 increased the expressions of brain TGF-ß/phosphorylated-Smad2/3 which was abrogated with the selective TGFß inhibitor SB431542. CONCLUSIONS: rCCL17-mediated Tregs recruitment may be a potential therapeutic strategy to promote M2 microglia/macrophages polarization and alleviate early brain injury following ICH.


Subject(s)
Brain Edema , Microglia , Mice , Male , Animals , Microglia/metabolism , Brain Edema/metabolism , Chemokines, CC/metabolism , Chemokines, CC/therapeutic use , T-Lymphocytes, Regulatory , Ligands , Macrophages/metabolism , Cerebral Hemorrhage/metabolism , Immunologic Factors , Disease Models, Animal , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/therapeutic use , Hematoma/metabolism
2.
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
3.
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
4.
J Orthop Res ; 37(8): 1723-1729, 2019 08.
Article in English | MEDLINE | ID: mdl-30977553

ABSTRACT

There is evidence that the application of mesenchymal stromal cells (MSCs) counteracts osteoarthritis (OA) progression. However, the prospect of extracting and expanding these cells might be limited. The aim of this study was to investigate whether hyaluronic acid (HA) supplemented with MSC-recruiting chemokine C-C motif ligand 25 (CCL25) can influence the natural course of spontaneous OA in the guinea pig. CCL25 concentration in synovial fluid (SF) was quantified with enzyme-linked immunosorbent assay. Boyden chamber cell migration assay was used to test CCL25-mediated migration of guinea pig MSC. Forty-nine 11-month-old male guinea pigs were divided into seven groups. The main treatments consisted of five intra-articular injections of HA in pure form and in combination with three doses of CCL25 (63, 693, and 6,993 pg) given at a weekly interval. The severity of cartilage damage was assessed by using a modified Mankin score. The measured average physiological concentration of CCL25 in SF of animals is 85 ± 39 pg/ml. MSC showed a 3.2-fold increase in cell migration at 1,000 nM CCL25 in vitro demonstrating the biological migratory activity of CCL25 on these cells. In vivo, treatment with HA alone did not reduce OA progression. Similarly, OA scores were not found significantly reduced after treatment with 63 pg CCL25 + HA. However, when compared to pure HA, treatment with 693 pg CCL25 + HA and 6,993 pg CCL25 + HA significantly reduced the OA score from 10.1 to 7.4 (-28%) and 8.4 (-20%), respectively. These data suggest that intra-articular injections of HA supplemented with CCL25 attenuates OA. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1723-1729, 2019.


Subject(s)
Arthritis, Experimental/drug therapy , Chemokines, CC/therapeutic use , Hyaluronic Acid/therapeutic use , Osteoarthritis, Knee/drug therapy , Viscosupplements/therapeutic use , Animals , Cartilage, Articular/drug effects , Cell Movement/drug effects , Chemokines, CC/metabolism , Chemokines, CC/pharmacology , Drug Evaluation, Preclinical , Guinea Pigs , Hyaluronic Acid/pharmacology , Injections, Intra-Articular , Male , Mesenchymal Stem Cells/drug effects , Synovial Fluid/metabolism , Viscosupplements/pharmacology
5.
Cancer Biol Ther ; 19(6): 475-483, 2018 06 03.
Article in English | MEDLINE | ID: mdl-29400599

ABSTRACT

Dendritic cell-cytokine-induced killer (DC-CIK) cell therapy has been experimentally implemented for enhancing anti-tumoral immunity in patients with hepatocellular carcinoma (HCC) undergoing postoperative transcatheter arterial chemoembolization (POTACE). We performed a retrospective study to evaluate the clinical efficacies of DC-CIK cell therapy and its correlations with several immune factors of the primary tumors. The overall survival time of HCC patients with HBV infection in the study group (POTACE plus DC-CIK cell therapy) was significantly longer than that of the control group (POTACE alone). The expression level of PD-L1 but not the tumor-infiltrated CD8 and CD4 T cells in the tumor tissues showed significant negative correlations with relapse-free survival (RFS) and overall survival (OS), which was also an independent prognostic factor for the five-years' suvival of patients with HCC receiving POTACE treatment. Furthermore, our study validated that PD-L1 expression was significantly inversely correlated with the survival time of HCC patients receiving POTACE plus DC-CIK cell therapy treatment. More importantly, DC-CIK cell therapy provided the best clinical benefits to HCC patients with the low PD-L1 expression receiving POTACE, which indicate that PD-L1 expression level can serve as a pivotal predictor for the therapeutic efficacy of DC-CIK cell therapy for HCC patients receiving POTACE treatment.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell- and Tissue-Based Therapy/methods , Chemoembolization, Therapeutic/methods , Chemokines, CC/therapeutic use , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/pathology , Chemokines, CC/pharmacology , Female , Humans , Liver Neoplasms/pathology , Male , Middle Aged , Postoperative Period , Prognosis , Retrospective Studies
6.
Stem Cells ; 34(4): 1112-22, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26763650

ABSTRACT

Mesenchymal stem cells (MSCs) possess the ability to migrate toward tumor sites and are regarded as promising gene delivery vehicles for cancer therapeutics. However, the factors that mediate this tropism have yet to be completely elucidated. In this study, through cytokine array analysis, chemokine CCL15 was found to be the most abundant protein differentially expressed in hepatocellular carcinoma (HCC) cell lines compared with a normal liver cell line. Serum CCL15 levels in HCC patients determined by enzyme linked immunosorbent assay were shown to be profoundly elevated compared with healthy controls. Immunohistochemical analysis indicated that CCL15 expression was much stronger in HCC tumor tissues than in adjacent nontumor tissues. Transwell migration assay suggested that CCL15 may be involved in chemotaxis of human MSCs (hMSCs) toward HCC in vitro and that this chemotactic effect of CCL15 is mediated via CCR1 receptors on hMSCs. Orthotopic animal models of HCC were established to investigate the role of CCL15 in hMSCs migration toward HCC in vivo. Both histological and flow cytometric analysis showed that significantly fewer hMSCs localized within 97H-CCL15-shRNA xenografts compared with 97H-green fluorescent protein xenografts after intravenous delivery. Finally, the possible effects of hMSCs on HCC tumor growth were also evaluated. Coculture experiments showed that hMSCs had no apparent effect on the proliferation of HCC cells in vitro In addition, systemic administration of hMSCs did not affect HCC tumor progression in vivo. Our data in this study help to elucidate the mechanism underlying the homing capacity of hMSCs toward HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Chemokines, CC/genetics , Gene Transfer Techniques , Liver Neoplasms/therapy , Macrophage Inflammatory Proteins/genetics , Mesenchymal Stem Cells/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement/genetics , Chemokines, CC/biosynthesis , Chemokines, CC/therapeutic use , Chemotaxis/genetics , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/genetics , Humans , Liver Neoplasms/genetics , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/therapeutic use , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/cytology , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Receptors, CCR1/biosynthesis , Receptors, CCR1/genetics , Xenograft Model Antitumor Assays
7.
Cytokine ; 59(1): 94-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22503614

ABSTRACT

The chemokine CCL28 participates in direct antimicrobial activities as well as homing of certain types of lymphocytes. The present study was conducted to harness these properties of the chemokine for the prevention of dental caries. The gene encoding CCL28 was transferred to salivary glands to enhance the production of this chemokine locally. First, a recombinant eukaryotic plasmid expressing CCL28 was constructed. Then, the CCL28 protein from 293 cells transfected with the recombinant plasmid was verified to inhibit the caries pathogen Streptococcus mutans (S. mutans) in a biofilm. Finally, the recombinant plasmid was retrogradely administered to the parotid glands of rats through the secretory ducts. The successful transfer of the gene encoding CCL28 to rat parotid acinar cells was confirmed by immunofluorescence and real-time PCR. Increases in both CCL28 and secretory IgA (SIgA) in the rat saliva were tested by ELISA. It was revealed that the CCL28 protein obtained from the study was able to strongly inhibit S. mutans living in biofilm in vitro. The delivery of the recombinant plasmid to the rat parotid glands was able to induce high levels of CCL28 and SIgA in saliva, and the increased levels of CCL28 and SIgA in saliva were maintained for 2 weeks. Notably, the dental plaque from the rats treated with the delivery of the recombinant plasmid in the study harbored significantly less S. mutans. These data indicated that the present strategy may hold hope for the effective prevention of dental caries.


Subject(s)
Chemokines, CC/genetics , Chemokines, CC/therapeutic use , Dental Caries/microbiology , Dental Caries/prevention & control , Gene Transfer Techniques , Salivary Glands/metabolism , Salivary Glands/microbiology , Animals , Anti-Infective Agents/metabolism , Chemokines, CC/metabolism , Dental Caries/therapy , Dental Plaque/microbiology , Eukaryotic Cells/metabolism , HEK293 Cells , Humans , Immunoglobulin A, Secretory/metabolism , Male , Plasmids/metabolism , Rats , Rats, Sprague-Dawley , Saliva/metabolism , Streptococcus mutans/cytology , Up-Regulation
8.
Leuk Res ; 35(9): 1254-60, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21295855

ABSTRACT

We have previously demonstrated that CCR9 plays a pivotal role in drug resistance and invasion in human acute T-lymphocytic leukemia (T-ALL). In this study, we investigated whether the MOLT4 cells, which naturally express CCR9 at high levels, can be successfully killed by the specific ligand, CCL25 fused to Pseudomonas exotoxin 38 (PE38) toxin. Our results demonstrated that CCL25-PE38 was able to specifically kill MOLT4 cells via apoptosis induction, and suppress the growth of CCR9(+) tumors. This work shows that CCR9 high-expressing human T-ALL cells can be successfully killed by delivering PE38 toxin fused to the ligand CCL25.


Subject(s)
ADP Ribose Transferases/therapeutic use , Apoptosis/drug effects , Bacterial Toxins/therapeutic use , Chemokines, CC/therapeutic use , Exotoxins/therapeutic use , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, CCR/metabolism , Virulence Factors/therapeutic use , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/pharmacology , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/pharmacology , Cells, Cultured , Chemokines, CC/chemistry , Chemokines, CC/pharmacology , Drug Evaluation, Preclinical , Exotoxins/chemistry , Exotoxins/pharmacology , Female , Humans , Mice , Mice, SCID , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Substrate Specificity , Virulence Factors/chemistry , Virulence Factors/pharmacology , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
9.
Allergy ; 63(10): 1317-23, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18782110

ABSTRACT

BACKGROUND: Modulation of leukocyte recruitment through blocking of chemokine receptors has been proposed as an attractive therapeutic strategy. We have previously demonstrated that n-Nonanoyl-CC chemokine ligand 14 (NNY-CCL14), a modified analog of the naturally occurring chemokine CCL14(9-74) internalizes and desensitizes human CCR3 resulting in the inactivation of eosinophils. However, inhibitory effects of NNY-CCL14 in murine models of allergic airway inflammation are assigned to its interaction with CCR1 and CCR5. AIM OF THE STUDY: As CCL2 and its receptor CCR2 have been shown to play important roles in the development of Th2 inflammation, we further evaluated the effects of NNY-CCL14 treatment on CCL2-mediated activation of CCR2. METHODS: Effects of NNY-CCL14 treatment were studied on cell lines transfected with human CCR2 and primary leukocytes. Functional effects were assessed by calcium efflux assays, flow cytometry and chemotaxis. RESULTS: Prestimulation with NNY-CCL14 desensitized CCR2-mediated responses to further stimulation with its selective ligand CCL2. No significant internalization of CCR2 was observed when the cells were stimulated with NNY-CCL14, even at concentrations eliciting maximal [Ca(2+)]i mobilization. Above all, NNY-CCL14 pretreatment blocked CCL2-induced chemotaxis of monocytes. CONCLUSIONS: This study demonstrates that NNY-CCL14 is a partial agonist of CCR2, inhibiting responses of monocytes to the CCR2-selective ligand CCL2. NNY-CCL14 attenuates CCR2-mediated responses by rapidly desensitizing the receptor and preventing chemotaxis, although it is able to induce calcium mobilization but does not lead to CCR2 internalization. Hence this study provides further insights into the possible mechanisms of action of NNY-CCL14, which interacts with multiple chemokine receptors inhibiting the migration and activation of different cell populations involved, thus acting as a potential therapeutic compound to alleviate allergic inflammation.


Subject(s)
Anti-Allergic Agents/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Chemokine CCL11/therapeutic use , Chemokines, CC/therapeutic use , Inflammation Mediators/therapeutic use , Receptors, CCR2/agonists , Respiratory Hypersensitivity/drug therapy , Animals , Anti-Allergic Agents/chemistry , Anti-Allergic Agents/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Line , Cell Migration Inhibition/drug effects , Cells, Cultured , Chemokine CCL11/chemistry , Chemokine CCL11/physiology , Chemokines, CC/chemistry , Chemokines, CC/physiology , Humans , Inflammation Mediators/physiology , Mice , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/biosynthesis , Respiratory Hypersensitivity/pathology
10.
Biotechnol Appl Biochem ; 48(Pt 2): 109-16, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17868025

ABSTRACT

CCL19 [chemokine (C-C motif) ligand 19; also known as MIP-3beta (macrophage inflammatory protein-3beta) or ELC (Epstein-Barr-virus-induced molecule 1 ligand chemokine)], one of the immunostimulatory cytokines, chemoattracts both DCs (dendritic cells) and T-lymphocytes. Adenoviral vector is one of the most used gene delivery vectors for cancer therapy because of its high gene-transfection efficiency. However, its wider application is limited, owing to immune responses that reduce transgene expression and decrease the efficacy of repeated administration. We constructed the recombinant replication deficient adenoviral vectors containing the CCL19 gene (Ad-CCL19) and combined them with PEG-PE [poly(ethylene glycol)-phosphatidylethanolamine]-modified cationic liposomes (Ad-CCL19/PEG-PE) for immunotherapy against murine fibrosarcoma. Although there were hardly any therapeutic differences between Ad-CCL19- and Ad-CCL19/PEG-PE-treated mice that were observed at the second administration, the final results demonstrated that Ad-CCL19/PEG-PE-treated mice survived much longer. The antitumour efficacy may be related to the high level of CCL19 after the final administration and lasting expression of IFN-gamma (interferon-gamma) and IL-12 (interleukin-12) in the Ad-CCL19/PEG-PE-treated group, which were measured by reverse-transcription PCR and ELISA. The results demonstrated that PEG-PE-cationic-liposome-conjugated adenovirus could prolong the expression of the therapeutic gene in vivo and may enhance the antitumour efficacy.


Subject(s)
Adenoviridae/genetics , Chemokines, CC/genetics , Chemokines, CC/therapeutic use , Gene Targeting/methods , Genetic Therapy/methods , Liposomes/chemistry , Lung Neoplasms/therapy , Adenoviridae/chemistry , Animals , Antineoplastic Agents/therapeutic use , Cations , Chemokine CCL19 , Female , Genetic Vectors/genetics , Lung Neoplasms/genetics , Mice , Mice, Inbred BALB C , Transfection/methods , Treatment Outcome
11.
Eur J Cardiothorac Surg ; 31(5): 773-8, 2007 May.
Article in English | MEDLINE | ID: mdl-17350855

ABSTRACT

OBJECTIVE: Local recurrence remains a major problem in the treatment of malignant pleural mesothelioma. The aim of the underlying study was to establish a standardised local recurrence model in rats which enables to study different intrapleural therapies. MATERIALS AND METHODS: Fifty microlitre containing 1 x 10(6) cells of a syngeneic rat malignant mesothelioma cell line (II-45), established from mesothelioma in Fischer 344 rats exposed to asbestos, were inoculated subpleurally via a left-sided thoracotomy. Tumour size was assessed 6 days later and the tumour nodule completely resected. Evaluation of recurrence at the resection site was performed after 10 days (n=6) and 6 days (n=6). The recurrent nodule was histopathologically confirmed. In a second experiment, this new recurrence model was evaluated for the effect of intrapleural therapy with different agents: 4 ml of cisplatin-solution (100mg(2)/kg BW), cisplatin combined with the fibrin-based sealant Vivostat, 4 ml taurolidine 2%, repeated injection of 1 microg of the chemokine CCL-19 at the tumour site and 4 ml povidone-iodine in a dilution 1:10. In a control group, the chest cavity was filled with 4 ml 0.9% NaCl. The primary endpoint was the extent of tumour recurrence. RESULTS: Six days after inoculation, all animals presented a standardised tumour nodule at the injection site of a mean diameter of 5.1 (+/-0.8)mm. Evaluation of the recurrence after 10 days showed a relapse directly at the resection site, but additional tumour nodules on the ipsi- and contralateral chest wall were found and histologically confirmed. The animals that were sacrificed 6 days after resection of the tumour nodule showed a recurrence only at the resection site with no macroscopic or microscopic evidence of other tumour. Resection of the tumour nodule combined with intrapleural application of the different agents lead to clear reduction of recurrence. The strongest effect was observed after intrapleural application of cisplatin-Vivostat with significant decrease of the longest, widest and thickest diameter of the recurrence. CONCLUSIONS: With this new recurrence model for investigation of malignant pleural mesothelioma in rats, we were able to investigate new intrapleural therapies after pneumonectomy. The intrapleural application of cisplatin-Vivostat significantly reduced the extent of local recurrence.


Subject(s)
Disease Models, Animal , Mesothelioma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Pleural Neoplasms/drug therapy , Animals , Anti-Infective Agents, Local/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Chemokine CCL19 , Chemokines, CC/therapeutic use , Cisplatin/therapeutic use , Combined Modality Therapy/methods , Fibrin Tissue Adhesive/therapeutic use , Male , Mesothelioma/surgery , Neoplasm Recurrence, Local/surgery , Pleural Neoplasms/surgery , Pneumonectomy/methods , Povidone-Iodine/therapeutic use , Rats , Rats, Inbred F344 , Taurine/analogs & derivatives , Taurine/therapeutic use , Thiadiazines/therapeutic use
12.
Mol Ther ; 13(1): 194-202, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16112911

ABSTRACT

Tyrosinase-related protein-2 (TRP2) is a weak antigen expressed in murine and human melanomas. Induction of antibody (Ab) response and T-cell immunity toward TRP2 with DNA plasmid vaccines has not been efficient to date. Recent studies have suggested that a chemokine ligand for the CCR7 (CCL21) present on T-cells and dendritic cells is important in activating and regulating immunity. We investigated the effectiveness of CCL21 as an adjuvant with an HVJ anionic liposomal TRP2 DNA (plasmid) vaccine to enhance anti-TRP2 Ab, cytokines, delayed-type hypersensitivity, T-cell responses, and tumor protection against B16 melanoma cells. Induction of anti-TRP2 immunity depended mainly on cell-mediated immunity, which was regulated by timing and route of CCL21 administration with DNA vaccine. The optimum protocol was to administer CCL21 im 24 h before DNA vaccine at the same vaccination site. Two vaccinations (prime/boost) were essential for induction of strong anti-TRP2 cell-mediated immunity. CCL21 administration 3 days before or 24 h after DNA vaccine, simultaneous with DNA vaccine, or at different sites (iv, opposite leg) was not effective. This study demonstrated that CCL21 was an effective adjuvant to enhance TRP2-specific immunity induced by a plasmid DNA cancer vaccine.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Cancer Vaccines/therapeutic use , Chemokines, CC/therapeutic use , Intramolecular Oxidoreductases/immunology , Melanoma, Experimental/therapy , Vaccines, DNA/therapeutic use , Adjuvants, Immunologic/administration & dosage , Animals , Antibody Formation , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Chemokine CCL21 , Chemokines, CC/administration & dosage , Chemokines, CC/immunology , Female , Immunity, Cellular , Liposomes , Lymph Nodes/immunology , Lymph Nodes/pathology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Spleen/immunology , Spleen/pathology , Transplantation, Heterologous , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
13.
Allergy ; 59(12): 1243-58, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15507091

ABSTRACT

The aim of this review is to give an overview of the role of chemokines, particularly ligands of the CC chemokine receptor CCR3, in allergic diseases and to show the new concept in the treatment of allergies using chemokine receptor antagonists. Allergic diseases such as allergic asthma, allergic rhinitis and atopic dermatitis are characterized by a complex interaction of different cell types and mediators. Among this, Th2 cells, mast cells, basophils and eosinophils are found in the inflamed tissue due to the attraction of chemokines. Of all the known chemokine receptors, the chemokine receptor CCR3 seems to play the major role in allergic diseases which is supported by the detection of this receptor on the cell types mentioned above. Therefore, academic and industrial research focus on compounds to block this receptor. To date, certain chemokine receptor antagonists derived from peptides and small molecules exist to block the chemokine receptor CCR3. However, the in vivo data about these compounds and the mechanisms of receptor interaction are poorly understood, as yet. For the development of additional chemokine receptor antagonists, more details about the interaction between the ligands and their receptors are required. Therefore, additional studies will lead to the identification of novel CCR3 chemokine receptor antagonists, which can be therapeutically used in allergic asthma, allergic rhinitis, and atopic dermatitis.


Subject(s)
Hypersensitivity/therapy , Receptors, Chemokine/antagonists & inhibitors , Amino Acid Sequence , Animals , Chemokine CCL11 , Chemokine CCL5/chemistry , Chemokine CCL5/immunology , Chemokine CCL5/therapeutic use , Chemokines/immunology , Chemokines, CC/chemistry , Chemokines, CC/immunology , Chemokines, CC/therapeutic use , Chemotactic Factors, Eosinophil/immunology , Glycosaminoglycans/immunology , Humans , Hypersensitivity/immunology , Ligands , Molecular Sequence Data , Receptors, CCR3 , Receptors, Chemokine/immunology
14.
J Leukoc Biol ; 76(1): 77-85, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15075363

ABSTRACT

The ideal vaccine carrier should be able to target antigen delivery and possibly recruit antigen-presenting cells (APC) and deliver an activation signal to promote adaptive immune responses. Ligands for chemokine receptors expressed on APC may be attractive candidates, as they can both target and attract APC. To investigate the requirement for APC recruitment, we used a pair of viral chemokines, agonist herpes simplex virus 8-derived macrophage inflammatory protein-I (vMIP-I) and antagonist MC148, which induce and suppress chemotaxis, respectively. Chemokine-antigen fusions efficiently delivered a model nonimmunogenic tumor antigen to APC for processing and presentation to antigen-specific T cells in vitro. Physical linkage of chemokine and antigen and specific binding of chemokine receptor by the fusion protein were required. Mice immunized with vMIP-I or MC148 fusion DNA vaccines elicited protection against tumor challenge. Therefore, vaccine efficacy depends primarily on the ability of the carrier to target antigen delivery to APC for subsequent processing and presentation, and chemotaxis directly induced by the chemokine moiety in the fusion may not be necessary.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Chemokines/therapeutic use , Drug Delivery Systems/methods , Genetic Therapy/methods , Lymphoma/prevention & control , Animals , Antigen-Presenting Cells/immunology , Cell Line, Tumor , Chemokines/genetics , Chemokines/immunology , Chemokines, CC/biosynthesis , Chemokines, CC/genetics , Chemokines, CC/immunology , Chemokines, CC/therapeutic use , Cloning, Molecular , Lymphoma/immunology , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/immunology , Macrophage Inflammatory Proteins/therapeutic use , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Vaccines, DNA/administration & dosage , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/therapeutic use
15.
J Biochem ; 136(6): 769-76, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15671487

ABSTRACT

Secondary lymphoid tissue chemokine (SLC) is a CC chemokine that plays an important role in leukocytes homing to lymphoid tissues. The ability of SLC to co-localize both T cells and dendritic cells formed the rationale to evaluate its utility in cancer immunotherapy. The in vivo antitumor effect of murine SLC (mSLC) has been well documented, but little is known about that of human SLC (hSLC). To investigate the antitumor efficiency in vivo of hSLC, the hSLC gene was artificially synthesized and induced to express as a soluble form in Escherichia coli. After purification, the purity of the recombinant human SLC (rhSLC) protein was above 95% by SDS-PAGE analysis. The K(d) of rhSLC binding to peripheral blood lymphocytes (PBLs) was 0.2186 +/- 0.02675 microM as assessed by FACS, and the maximal chemotactic index of rhSLC was 9.49 at 100 nM as assessed by in vitro chemotaxis assay. Then genomic sequences of hSLC and mSLC, and of human CCR7 (hCCR7) and murine CCR7 (mCCR7), the receptor for SLC, were aligned. It was found that hSLC and mSLC share 70.72% identity and hCCR7 and mCCR7share 86.77% identity. Furthermore, we found that rhSLC could chemoattract murine peripheral blood mononuclear cells (PBMCs) in vitro. On the basis of these facts, immune competent mice inoculated with S180 sarcoma cells were chosen as an in vivo model. Intratumoral injections of rhSLC inhibited tumor growth and increased survival. These findings suggest that, despite its incapability to bind to either human or murine CXCR3, which is related to angiostasis, rhSLC can induce an antitumor response in vivo by another route. This report proves that rhSLC has a potent tumor-inhibition ability that makes it a promising candidate agent in cancer immunotherapy.


Subject(s)
Angiogenesis Inhibitors/immunology , Angiogenesis Inhibitors/therapeutic use , Chemokines, CC/immunology , Chemokines, CC/therapeutic use , Genetic Therapy , Sarcoma, Experimental/prevention & control , Adult , Angiogenesis Inhibitors/metabolism , Animals , Chemokine CCL21 , Chemokines, CC/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , In Vitro Techniques , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, SCID , Receptors, Chemokine/metabolism , Recombinant Proteins/therapeutic use , Research , Sarcoma, Experimental/immunology , Sarcoma, Experimental/metabolism , Survival Rate , T-Lymphocytes/immunology , Tumor Cells, Cultured
17.
Cancer Chemother Pharmacol ; 50(2): 163-6, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12172984

ABSTRACT

PURPOSE: Chemokines are a family of small proteins that regulate leukocyte infiltration into inflamed tissue and play key roles in the pathogenesis of many diseases. Some chemokines can also reversibly inhibit the proliferation of hematopoietic progenitors. We have previously found that the chemokine CCL21 (Exodus-2/SLC/6Ckine/TCA4) is a potent inhibitor of the proliferation of normal hematopoietic progenitors. In this study we sought to determine whether this inhibition of proliferation could be therapeutically exploited by protecting normal marrow progenitors from the cytotoxicity of the S phase-active chemotherapeutic agent Ara-C. METHODS: Untreated and CCL21-pretreated mice were given doses of Ara-C that are toxic to marrow myeloid progenitors. The recovery of these myeloid progenitors was analyzed by colony formation assays. RESULTS: It was found that pretreatment with small doses of CCL21 prevented the death of normal murine marrow progenitors from the toxic effects of Ara-C. CONCLUSIONS: The chemokine CCL21 may be able to prevent Ara-C myelosuppression during acute leukemia induction chemotherapy, and thereby decrease morbidity and mortality of such therapy, and shorten hospital stays.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Bone Marrow Diseases/prevention & control , Bone Marrow/drug effects , Chemokines, CC/therapeutic use , Cytarabine/antagonists & inhibitors , Hematopoietic Stem Cells/drug effects , Animals , Bone Marrow Diseases/chemically induced , Cell Division/drug effects , Chemokine CCL21 , Colony-Forming Units Assay , Cytarabine/toxicity , Drug Evaluation, Preclinical , Mice , S Phase/drug effects
18.
Cancer Immunol Immunother ; 49(11): 587-92, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11225989

ABSTRACT

The recently described CC chemokine, 6C-kine, is unique in that it contains -six rather than the usual four conserved cysteines typical of this family. Furthermore, murine 6C-kine binds to one of the CXC chemokine receptors CXCR3, in addition to its other known receptor CCR7. We have shown that two other ligands of CXCR3, IP-10 and MIG, are potent inhibitors of tumor growth in severe combined immunodeficiency (SCID) mice. We postulated that murine 6C-kine may also inhibit tumor growth via inhibition of angiogenesis in this model. SCID mice (n = 6 per group) inoculated with A549 human lung cancer cells were treated with either 6C-kine (100 ng intra-tumor injection every other day) or control protein for 8 weeks. Tumors from murine 6C-kine-treated mice (288 +/- 26 mm3) were significantly smaller than tumors from control treated mice (788 +/- 156 mm3, P = 0.005). Additionally, murine 6C-kine reduced metastases compared with controls (0.5 +/- 0.3 vs 3.0 +/- 1.2 metastases per animal, P = 0.05). Tumor vascularity (as assessed by vessel density counting) was reduced in murine 6C-kine-treated mice compared with controls. Murine 6C-kine had no direct effect on proliferation of A549 cells, and there were no differences in the infiltration of leukocyte sub-populations, assessed by flow cytometry, in the treatment groups. Interestingly, human 6C-kine, unlike murine 6C-kine, does not bind CXCR3 and had no anti-tumor effect in the same model. These data suggest that murine 6Ckine has anti-tumor effects independent of its leukocyte-recruiting activity. Furthermore, while not confirmatory, these data lend further support to the fact that CXCR3 may be the receptor for angiostatic CXC chemokines.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Chemokines, CC/therapeutic use , Lung Neoplasms/drug therapy , Neoplasms, Experimental/drug therapy , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Division/drug effects , Chemokine CCL21 , Chimera , Female , Humans , Leukocytes/immunology , Lung Neoplasms/pathology , Mice , Mice, SCID , Neoplasm Metastasis , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/drug therapy , Receptors, CXCR3 , Receptors, Chemokine/physiology , Tumor Cells, Cultured
19.
Infect Immun ; 68(11): 6108-14, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11035713

ABSTRACT

Previous studies have suggested that the C-C chemokine C10 is involved in the chronic stages of host defense reactions. The present study addressed the role of C10 in a murine model of septic peritonitis, induced by cecal ligation and puncture (CLP). Unlike other C-C chemokines, C10 levels in the peritoneal wash were increased approximately 30-fold above baseline levels at 48 h after CLP surgery. Immunoneutralization of peritoneal C10 levels with polyclonal anti-C10 antiserum during CLP-induced peritonitis negatively impacted mouse survival over 4 days. In contrast, when 500 ng of recombinant murine C10 was administered immediately after CLP surgery, the 4-day survival rate increased from 20% to over 60%. The C10 therapy appeared to facilitate a rapid and significant enhancement of the levels of tumor necrosis factor alpha (TNF-alpha) and monocyte chemoattractant protein-1 (MCP-1) and a later increase in interleukin-13 (IL-13) levels in the peritoneal cavity. In vitro studies showed that the combination of IL-1beta and C10 markedly augmented TNF-alpha synthesis by peritoneal macrophages and that C10 synthesis was induced in these cells following their exposure to IL-13. At 24 h after CLP surgery, only 25% of C10-treated mice were bacteremic versus 85% of the control group that exhibited dissemination of bacteria into the circulation. The lack of bacteremia in C10-treated mice appeared to be related, in part, to in vitro evidence that C10 significantly enhanced the bacterial phagocytic activity of peritoneal macrophages. In addition, in vivo evidence suggested that C10 therapy significantly reduced the amount of material that leaked from the damaged gut. Taken together, the results of this study demonstrate that the C10 chemokine rapidly promotes disease resolution in the CLP model through its direct effects on the cellular events critically involved in host defense during septic peritonitis.


Subject(s)
Bacteremia/immunology , Chemokines, CC/physiology , Animals , Bacteremia/mortality , Bacteremia/therapy , Chemokine CCL2/biosynthesis , Chemokines, CC/therapeutic use , Female , Immune Sera/immunology , Interleukin-1/pharmacology , Interleukin-13/biosynthesis , Interleukin-13/pharmacology , Mice , Phagocytosis , Rabbits , Recombinant Proteins/therapeutic use , Tumor Necrosis Factor-alpha/biosynthesis
20.
J Immunol ; 165(4): 1992-2000, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10925282

ABSTRACT

Mouse 6Ckine/SLC (secondary lymphoid tissue chemokine) is a chemotactic factor for dendritic cells, T cells, and NK cells in vitro. In addition, mouse 6Ckine/SLC interacts with the chemokine receptor CXCR3, as do several chemokines with antiangiogenic properties. These dual properties of mouse 6Ckine/SLC were tested for the induction of an antitumor response by transducing the C26 colon carcinoma tumor cell line with a cDNA encoding mouse 6Ckine/SLC. The C26-6CK-transduced cells showed reduced tumorigenicity in immunocompetent or in nude mice. Part of this effect was likely due to angiostatic mechanisms as shown by immunohistochemistry and Matrigel assay. C26-6CK tumors were also heavily infiltrated with leukocytes, including granulocytes, dendritic cells, and CD8+ T cells. In vivo, anti-CD8 treatment increased the tumorigenicity of the C26-6CK tumor cells, and tumor-infiltrating CD8+ T cells had the phenotype of memory effector cells, suggesting the induction of cytotoxic tumor-specific T lymphocytes. On the other hand, anti-asialo-GM1 depletion also increased the tumorigenicity of C26-6CK cells, supporting the participation of NK cells. Finally, tumor-infiltrating dendritic cells had the phenotype and functional features of immature dendritic cells. Overall, these results suggest that mouse 6Ckine/SLC has strong antitumor effects by inducing both angiostatic, CD8+ T cell-mediated, and possibly NK-mediated tumor resistance mechanisms.


Subject(s)
Angiogenesis Inhibitors/immunology , Angiogenesis Inhibitors/therapeutic use , Chemokines, CC/immunology , Chemokines, CC/therapeutic use , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/immunology , Angiogenesis Inhibitors/administration & dosage , Animals , Cell Division/genetics , Cell Division/immunology , Cell Movement/immunology , Chemokine CCL21 , Chemokines, CC/administration & dosage , Chemokines, CC/genetics , Cytokines/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Female , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Immunophenotyping , Leukocytes/immunology , Leukocytes/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , RNA, Messenger/biosynthesis , Receptors, CCR7 , Receptors, CXCR3 , Receptors, Chemokine/biosynthesis , Receptors, Chemokine/genetics , Tumor Cells, Cultured/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...