Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Wound Repair Regen ; 28(1): 39-48, 2020 01.
Article in English | MEDLINE | ID: mdl-31587435

ABSTRACT

To study the effects of mir-27b on angiogenesis and fibroblast activation and to explore its further mechanism. Humanmicrovascular endothelial cell (HMEC)-1 and humannormal skin fibroblast (BJ) cells were treated with mir-27b inhibitor negative control reagent, mir-27b inhibitor, LY294002, and mir-27b inhibitor + LY294002, respectively. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was used to detect the T-cell proliferation. The migration ability was detected by Scratch assays. The angiogenesis of HMEC-1 cells was observed by in vitro tube formation assay. The mRNA and protein expression of vascular endothelial growth factor (VEGF) in HMEC-1 cells and the mRNA and protein expression of collagen I, collagen III, α-SMA, and MMP1 in BJ cells were detected by quantitativereal-time polymerase chain reaction (qRT-PCR) and Western blot, respectively. Meanwhile, the PI3K/protein kinase B (AKT) pathway-related proteins were also detected by Western blot. The proliferation, migration, angiogenesis, the mRNA and protein expression of VEGF and the protein expression of p-PI3K and p-AKT in HMEC-1 cells were increased after treated with mir-27b inhibitor. Meanwhile, the proliferation, migration, and the protein expression of collagen I, collagen III, α-SMA, MMP1, p-PI3K, and p-AKT in BJ cells were increased after treated with mir-27b inhibitor. However, the angiogenesis and fibroblast activation of mir-27b inhibitor was reversed by LY294002, and the activate effect to PI3K/AKT pathway was also inhibited. Down-regulation of mir-27b could promote angiogenesis and fibroblast activation, and its mechanism is related to activate PI3K/AKT signaling pathway.


Subject(s)
Cell Proliferation/genetics , Endothelial Cells/metabolism , Fibroblasts/metabolism , Neovascularization, Physiologic/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Actins/drug effects , Actins/genetics , Actins/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Chromones/pharmacology , Collagen Type I/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type III/drug effects , Collagen Type III/genetics , Collagen Type III/metabolism , Down-Regulation , Endothelial Cells/drug effects , Fibroblasts/drug effects , Humans , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Morpholines/pharmacology , Neovascularization, Physiologic/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Signal Transduction , Skin/cytology , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Wound Healing
2.
Bioorg Chem ; 93: 103323, 2019 12.
Article in English | MEDLINE | ID: mdl-31586713

ABSTRACT

New 1,3,4-thiadiazine-thiourea derivatives have been synthesized. All the synthesized compounds were examined for in vitro cytotoxic activity against Non-Small Cell Lung Cancer (NSCLC) cell line A549, using MTT bioassay. Compounds 5d, 5i, 5j showed the highest cytotoxic activity with IC50 values of 0.27 ±â€¯0.01, 0.30 ±â€¯0.02, and 0.32 ±â€¯0.012 µM respectively with sorafenib as reference (IC50 3.85 ±â€¯0.27 µM). These compounds were chosen for further investigations against various biological targets known to play roles in NSCLC specifically: vascular endothelial growth factor receptor 2 (VEGFR2), B-RAF and matrix metalloproteinase 9 (MMP9). Encouraging results were exhibited by the three compounds against the selected targets. Compound 5j was specially promising as it exhibited inhibitory activity of VEGFR2 close to sorafenib (IC50 0.11 ±â€¯0.01 µM), most potent B-RAF activity inhibition (IC50 0.178 ±â€¯0.004 µM) and MMP9 inhibition (IC50 0.08 ±â€¯0.004 µM). Moreover, cell cycle analysis of A549 cells treated with 5j exhibited cell cycle arrest at G2-M phase and pro-apoptotic activity as indicated by Annexin V-FITC staining. Also, it reflected antinvasive and antimigration properties to A549 cells. Additionally, docking study of 5j on VEGFR2, B-RAF and MMP9 revealed that it binds to the target enzymes in a similar way as the co-crystallized ligand. The three compounds exhibited significantly high selectivity to A549 cancer cells against the normal human fetal lung fibroblast cell line WI-38 with higher selectivity index compared to sorafenib (5d IC50 136.76 ±â€¯2.38 µM, SI = 506.52; 5i IC50 89.20 ±â€¯2.11 µM, SI = 297.33; 5j IC50 79.60 ±â€¯3.8 µM, SI = 248.75; sorafenib IC50 30.32 ±â€¯2.41 µM, SI = 7.88). In conclusion, compounds 5d, 5i and 5j, specially 5j are promising anticancer agents targeting important pathways in NSCLC and warrant further preclinical and clinical trials.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Design , Thiadiazines/chemistry , Thiadiazines/pharmacology , A549 Cells , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Drug Screening Assays, Antitumor , Humans , Lung Neoplasms/pathology , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 9/drug effects , Molecular Docking Simulation , Neoplasm Metastasis/prevention & control , Proto-Oncogene Proteins B-raf/drug effects , Thiadiazines/chemical synthesis , Vascular Endothelial Growth Factor Receptor-2/drug effects
3.
Clin Oral Investig ; 22(3): 1243-1252, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28965247

ABSTRACT

OBJECTIVES: This article evaluated the drug loading, release kinetics, and matrix metalloproteinase (MMP) inhibition of doxycycline (DOX) released from DOX-loaded nanotube-modified adhesives. DOX was chosen as the model drug, since it is the only MMP inhibitor approved by the U.S. Food and Drug Administration. MATERIALS AND METHODS: Drug loading into the nanotubes was accomplished using DOX solution at distinct concentrations. Increased concentrations of DOX significantly improved the amount of loaded DOX. The modified adhesives were fabricated by incorporating DOX-loaded nanotubes into the adhesive resin of a commercial product. The degree of conversion (DC), Knoop microhardness, DOX release kinetics, antimicrobial, cytocompatibility, and anti-MMP activity of the modified adhesives were investigated. RESULTS: Incorporation of DOX-loaded nanotubes did not compromise DC, Knoop microhardness, or cell compatibility. Higher concentrations of DOX led to an increase in DOX release in a concentration-dependent manner from the modified adhesives. DOX released from the modified adhesives did not inhibit the growth of caries-related bacteria, but more importantly, it did inhibit MMP-1 activity. CONCLUSIONS: The loading of DOX into the nanotube-modified adhesives did not compromise the physicochemical properties of the adhesives and the released levels of DOX were able to inhibit MMP activity without cytotoxicity. CLINICAL SIGNIFICANCE: Doxycycline released from the nanotube-modified adhesives inhibited MMP activity in a concentration-dependent fashion. Therefore, the proposed nanotube-modified adhesive may hold clinical potential as a strategy to preserve resin/dentin bond stability.


Subject(s)
Anti-Bacterial Agents/chemistry , Doxycycline/chemistry , Matrix Metalloproteinase 1/drug effects , Nanotubes/chemistry , Resin Cements/chemistry , Cell Culture Techniques , Chromatography, High Pressure Liquid , Hardness , Materials Testing , Matrix Metalloproteinase Inhibitors/chemistry
4.
Rheumatology (Oxford) ; 56(8): 1417-1427, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28431044

ABSTRACT

Objective: To explore the role of leonurine in the regulation of synovial inflammation and joint destruction inRA. Methods: Fibroblast-like synoviocytes were isolated from synovial tissue from RA patients. Pro-inflammatory cytokine and MMP expression was evaluated using real-time PCR and a cytometric bead array. Cell migration and invasion in vitro were measured using the Boyden chamber method and the scratch assay, respectively. Protein expression was measured by western blotting. Nuclear factor kappa B (NF-κB) nuclear translocation was detected by immunofluorescence. The in vivo effect of leonurine was evaluated in mice with CIA. Results: Leonurine treatment significantly decreased the production of pro-inflammatory cytokines (IL-1ß, IL-6, IL-8 and TNFα) and MMPs (MMP-1 and MMP-3) and suppressed the migration and invasion of RA fibroblast-like synoviocytes. The molecular analysis revealed that leonurine impaired TNFα-induced NF-κB signalling by inhibiting the phosphorylation and degradation of inhibitor of NF-κB alpha (IκBα) and subsequently preventing the nuclear translocation of the NF-κB p65 subunit. Leonurine also inhibited the p38 and Jun N-terminal kinase mitogen-activated protein kinases signalling pathways without affecting ERK signalling. Intraperitoneal injection of leonurine reduced synovial inflammation, joint destruction and the serum IL-1ß, IL-6 and TNFα levels in mice with CIA. Conclusion: Our findings show that leonurine reduces synovial inflammation and joint destruction in RA through the NF-κB and mitogen-activated protein kinases pathways. Leonurine has potential as a therapeutic agent for RA.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Gallic Acid/analogs & derivatives , Adult , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Cytokines/drug effects , Female , Fibroblasts/metabolism , Gallic Acid/pharmacokinetics , Humans , Inflammation Mediators/metabolism , Male , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 3/drug effects , Mice , Middle Aged , Mitogen-Activated Protein Kinases/drug effects , Protein Transport/drug effects , Signal Transduction/drug effects , Synovial Membrane/drug effects , Synoviocytes/drug effects , Synoviocytes/pathology , Transcription Factor RelA/drug effects
5.
Rheumatology (Oxford) ; 56(3): 351-361, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27940589

ABSTRACT

Objective: A crucial feature of OA is cartilage degradation. This process is mediated by pro-inflammatory cytokines, among other factors, via induction of matrix-degrading enzymes. Interleukin 37 (IL37) is an anti-inflammatory cytokine and is efficient in blocking the production of pro-inflammatory cytokines during innate immune responses. We hypothesize that IL37 is therapeutic in treating the inflammatory cytokine cascade in human OA chondrocytes and can act as a counter-regulatory cytokine to reduce cartilage degradation in OA. Methods: Human OA cartilage was obtained from patients undergoing total knee or hip arthroplasty. Immunohistochemistry was applied to study IL37 protein expression in cartilage biopsies from OA patients. Induction of IL37 expression by IL1ß, OA synovium-conditioned medium and TNFα was investigated in human OA chondrocytes. Adenoviral overexpression of IL37 followed by IL1ß stimulation was performed to investigate the anti-inflammatory potential of IL37. Results: IL37 expression was detected in cartilage biopsies of OA patients and induced by IL1ß. After IL1ß stimulation, increased IL1ß, IL6 and IL8 expression was observed in OA chondrocytes. Elevated IL37 levels diminished the IL1ß-induced IL1ß , IL6 and IL8 gene levels and IL1ß and IL8 protein levels. In addition to the reduction in pro-inflammatory cytokine expression, IL37 reduced MMP1 , MMP3 , MMP13 and disintegrin and metalloproteinase with thrombospondin motifs 5 gene levels and MMP3 and MMP13 protein levels. Conclusion: IL37 is induced by IL1ß, and IL37 itself reduced IL1ß, IL6 and IL8 production, indicating that IL37 is able to induce a counter-regulatory anti-inflammatory feedback loop in chondrocytes. In addition, IL37 dampens catabolic enzyme expression. This supports IL37 as a potential therapeutic target in OA.


Subject(s)
Chondrocytes/metabolism , Interleukin-1/metabolism , Interleukin-1beta/pharmacology , Osteoarthritis , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Adenoviridae , Blotting, Western , Chondrocytes/drug effects , Disintegrins/drug effects , Disintegrins/genetics , Disintegrins/metabolism , Humans , Immunohistochemistry , Interleukin-1/genetics , Interleukin-1beta/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/drug effects , Interleukin-8/genetics , Interleukin-8/metabolism , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 13/drug effects , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , RNA, Messenger/drug effects , Real-Time Polymerase Chain Reaction
6.
Orthod Craniofac Res ; 20(1): 30-34, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28102013

ABSTRACT

OBJECTIVES: The aim of this study was to analyze MMP-1 transcript levels in periodontal tissues of rats that underwent orthodontic treatment using potassium diclofenac and dexamethasone at different stages of tooth movement. SETTING AND SAMPLE POPULATION: The sample comprised of ninety male Wistar rats. MATERIAL AND METHODS: A closed nickel-titanium coil spring was used to apply a force of 50 cN to move the maxillary right first molars mesially. One group received daily doses of 0.9% saline solution, the second group received daily doses of 5 mg/kg potassium diclofenac, and the third group received daily doses of 0.5 mg/kg dexamethasone. Tooth movement was observed on days 0, 1, 3, 7, and 14. MMP-1 transcript levels were evaluated by real-time polymerase chain reaction and the results were compared between groups by three-way ANOVA, with a significance level of 0.05. RESULTS: Transcript levels increased in groups that received the coil spring treatment on all days of the experiment. MMP-1 expression was found to be decreased in groups treated with potassium diclofenac and dexamethasone compared to that in the control group, on days 1, 3, 5, and 7. CONCLUSIONS: The application of orthodontic forces significantly increased MMP-1 transcript levels. The use of anti-inflammatory drugs may have an inhibitory effect on MMP-1 expression.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Diclofenac/pharmacology , Matrix Metalloproteinase 1/drug effects , Tooth Movement Techniques , Animals , Male , Matrix Metalloproteinase 1/metabolism , Periodontium/drug effects , Periodontium/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats, Wistar
7.
Int J Cosmet Sci ; 39(2): 133-140, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27502959

ABSTRACT

OBJECTIVE: Licoricidin is an isoflavonoid isolated from Glycyrrhiza uralensis Fisher. In this study, we investigated the effects of licoricidin on photoaging of UVA-irradiated human dermal fibroblasts (HDFs). METHODS: In vitro reactive oxygen species (ROS) scavenging activity, cellular protective effect and inhibition of elastase activity was determined by Fe3+ -EDTA/H2 O2 systems, photohaemolysis and elastase activity assay, respectively. Anti-oxidative capacity of the compound was evaluated by fluorescent ELISA and 2', 7'-dichlorofluorescin-diacetate (DCF-DA) assay. The expression of protein and phosphorylation was examined using Western blot. RESULTS: The ROS scavenging activity (OSC50 ) of licoricidin was 2.77 µM. It was 3.1-fold higher than that of L-ascorbic acid. Its protective effects were confirmed in a study of 1 O2 -induced cellular damage to human erythrocytes. The τ50 value of 10 µM of licoricidin was 71.0 min; this was markedly higher than that obtained with α-tocopherol (37.0 min). The elastase inhibitory activity of licoricidin (IC50 of 61.2 µM) was 2.1-fold more potent than that of oleanolic acid. Licoricidin markedly reduced the UVA-induced intracellular ROS in a concentration-dependent manner. Western blot revealed that licoricidin attenuated the UVA-dependent induction of MMP-1 protein. Mechanistically, this appeared to be due to licoricidin-dependent inhibition of mitogen-activated protein kinases (MAPK) phosphorylation, which resulted in decreased c-Jun activation and reduced c-Jun and c-Fos expression. CONCLUSION: Licoricidin blocks UVA-induced photoaging via ROS scavenging. This activity converges to limit the activity of MMP-1. These data suggest that licoricidin may be considered as an active ingredient in new topically applied anti-ageing formulations.


Subject(s)
Benzopyrans/pharmacology , Glycyrrhiza uralensis/chemistry , Skin/drug effects , Ultraviolet Rays , Benzopyrans/isolation & purification , Enzyme Activation/radiation effects , Enzyme Induction , Fibroblasts/drug effects , Humans , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 1/drug effects , Mitogen-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Skin/cytology , Skin/metabolism , Skin/radiation effects , Skin Aging
8.
Pharmazie ; 71(2): 94-100, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27004374

ABSTRACT

The vascular remodeling process plays an important role in the pathology of hypoxia-induced pulmonary hypertension, and it includes cell proliferation, cell motility, cell synthesis and collagen coagulation. Due to their proliferation and synthesis ability, the adventitial fibroblasts are thought to be critical in the vascular remodeling process initiated in response to hypoxia. However, the factors driving hypoxia-induced fibroblast proliferation and synthesis have yet to be elucidated, and the treatment regimens to treat hypoxia remain ineffective. As forthis study, its purpose was to examine the effects exerted by SB-431542, a small-molecule antagonist of transforming growth factor-ß-receptor, on the proliferation, synthesis and collagen coagulation in cultured adventitial fibroblasts. Another aim of this study was to assess the inhibitory ability of SB-431542 on pulmonary vascular remodeling in chronic hypoxia in vivo.The cell morphology and proliferation of cultured adventitial fibroblasts was assessed by laser confocal microscopy and the MTT assay, respectively. Additionally, collagen synthesis was determined by hydroxyproline chromatography, while the expression of cytokines in adventitial fibroblasts and lung tissues was evaluated by immunohistochemical and reverse transcription PCR analyses. The results indicated that the exposure of cultured fibroblasts to 1% oxygen led to the up regulation of cell proliferation, cell synthesis. In addition, increased expression of cytokines and collagen was detected in vivo in the pulmonary artery adventitia of rats exposed to chronic hypoxia. Conversely, SB-431542 inhibited fibroblast proliferation and synthesis in the process of hypoxia-induced pulmonary hypertension (P < 0.01). Thus, the results suggested that by reducing cell proliferation, cell synthesis of vascular adventitia, small molecule inhibitors of the TGF-ß1 receptors may offer a novel therapy for pulmonary hypertension.


Subject(s)
Benzamides/pharmacology , Dioxoles/pharmacology , Fibroblasts/drug effects , Hypoxia/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pulmonary Artery/pathology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Actins/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Cells, Cultured , Collagen/biosynthesis , Cytokines/biosynthesis , Down-Regulation/drug effects , Hydroxyproline/metabolism , Male , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/biosynthesis , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I
9.
Ann Rheum Dis ; 74(1): 303-10, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24285492

ABSTRACT

OBJECTIVES: Due to their role in inflammatory metabolic diseases, we hypothesised that free fatty acids (FFA) are also involved in inflammatory joint diseases. To test this hypothesis, we analysed the effect of FFA on synovial fibroblasts (SF), human chondrocytes and endothelial cells. We also investigated whether the toll-like receptor 4 (TLR4), which can contribute to driving arthritis, is involved in FFA signalling. METHODS: Rheumatoid arthritis SF, osteoarthritis SF, psoriatic arthritis SF, human chondrocytes and endothelial cells were stimulated in vitro with different FFA. Immunoassays were used to quantify FFA-induced protein secretion. TLR4 signalling was inhibited extracellularly and intracellularly. Fatty acid translocase (CD36), responsible for transporting long-chain FFA into the cell, was also inhibited. RESULTS: In rheumatoid arthritis synovial fibroblasts (RASF), FFA dose-dependently enhanced the secretion of the proinflammatory cytokine IL-6, the chemokines IL-8 and MCP-1, as well as the matrix-degrading enzymes pro-MMP1 and MMP3. The intensity of the response was mainly dependent on the patient rather than on the type of disease. Both saturated and unsaturated FFA showed similar effects on RASF, while responses to the different FFA varied for human chondrocytes and endothelial cells. Extracellular and intracellular TLR4 inhibition as well as fatty acid transport inhibition blocked the palmitic acid-induced IL-6 secretion of RASF. CONCLUSIONS: The data show that FFA are not only metabolic substrates but may also directly contribute to articular inflammation and degradation in inflammatory joint diseases. Moreover, the data suggest that, in RASF, FFA exert their effects via TLR4 and require extracellular and intracellular access to the TLR4 receptor complex.


Subject(s)
Arthritis, Psoriatic/immunology , Arthritis, Rheumatoid/immunology , Chondrocytes/immunology , Endothelial Cells/immunology , Fatty Acids, Nonesterified/immunology , Fibroblasts/immunology , Inflammation Mediators/immunology , Osteoarthritis/immunology , Signal Transduction/immunology , CD36 Antigens/drug effects , CD36 Antigens/metabolism , Chemokine CCL2/drug effects , Chemokine CCL2/immunology , Chondrocytes/drug effects , Endothelial Cells/drug effects , Fatty Acids, Nonesterified/pharmacology , Fibroblasts/drug effects , Humans , Interleukin-6/immunology , Interleukin-8/drug effects , Interleukin-8/immunology , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/immunology , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/immunology , Signal Transduction/drug effects , Synovial Membrane/cytology , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/metabolism
10.
Ann Rheum Dis ; 74(6): 1311-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25398374

ABSTRACT

OBJECTIVE: Tofacitinib is an oral Janus kinase (JAK) inhibitor for the treatment of rheumatoid arthritis (RA). The pathways affected by tofacitinib and the effects on gene expression in situ are unknown. Therefore, tofacitinib effects on synovial pathobiology were investigated. METHODS: A randomised, double-blind, phase II serial synovial biopsy study (A3921073; NCT00976599) in patients with RA with an inadequate methotrexate response. Patients on background methotrexate received tofacitinib 10 mg twice daily or placebo for 28 days. Synovial biopsies were performed on Days -7 and 28 and analysed by immunoassay or quantitative PCR. Clinical response was determined by disease activity score and European League Against Rheumatism (EULAR) response on Day 28 in A3921073, and at Month 3 in a long-term extension study (A3921024; NCT00413699). RESULTS: Tofacitinib exposure led to EULAR moderate to good responses (11/14 patients), while placebo was ineffective (1/14 patients) on Day 28. Tofacitinib treatment significantly reduced synovial mRNA expression of matrix metalloproteinase (MMP)-1 and MMP-3 (p<0.05) and chemokines CCL2, CXCL10 and CXCL13 (p<0.05). No overall changes were observed in synovial inflammation score or the presence of T cells, B cells or macrophages. Changes in synovial phosphorylation of signal transducer and activator of transcription 1 (STAT1) and STAT3 strongly correlated with 4-month clinical responses (p<0.002). Tofacitinib significantly decreased plasma CXCL10 (p<0.005) at Day 28 compared with placebo. CONCLUSIONS: Tofacitinib reduces metalloproteinase and interferon-regulated gene expression in rheumatoid synovium, and clinical improvement correlates with reductions in STAT1 and STAT3 phosphorylation. JAK1-mediated interferon and interleukin-6 signalling likely play a key role in the synovial response. TRIAL REGISTRATION NUMBER: NCT00976599.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Janus Kinase 1/drug effects , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , RNA, Messenger/drug effects , STAT Transcription Factors/drug effects , Synovial Membrane/drug effects , Adult , Aged , Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/metabolism , Chemokines/drug effects , Chemokines/genetics , Chemokines/metabolism , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Janus Kinase 1/metabolism , Male , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Methotrexate/therapeutic use , Middle Aged , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , RNA, Messenger/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Synovial Membrane/metabolism , Treatment Outcome
11.
Rheumatology (Oxford) ; 54(6): 1087-92, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25389358

ABSTRACT

OBJECTIVE: Glucocorticoids are powerful anti-inflammatory compounds that also induce the expression of leptin and leptin receptor (Ob-R) in synovial fibroblasts through TGF-ßsignalling and Smad1/5 phosphorylation. Compound A (CpdA), a selective glucocorticoid receptor agonist, reduces inflammation in murine arthritis models and does not induce diabetes or osteoporosis, thus offering an improved risk:benefit ratio in comparison with glucocorticoids. Due to the detrimental role of leptin in OA pathogenesis, we sought to determine whether CpdA also induced leptin and Ob-R protein expression as observed with prednisolone. METHODS: Human synovial fibroblasts and chondrocytes were isolated from the synovium and cartilage of OA patients after joint surgery. The cells were treated with prednisolone, TGF-ß1, TNF-α and/or CpdA. Levels of leptin, IL-6, IL-8, MMP-1 and MMP-3 were measured by ELISA and expression levels of Ob-R phospho-Smad1/5, phospho-Smad2, α-tubulin and glyceraldehyde 3-phosphate dehydrogenase were analysed by western blotting. RESULTS: CpdA, unlike prednisolone, did not induce leptin secretion or Ob-R protein expression in OA synovial fibroblasts. Moreover, CpdA decreased endogenous Ob-R expression and down-regulated prednisolone-induced leptin secretion and Ob-R expression. Mechanistically, CpdA, unlike prednisolone, did not induce Smad1/5 phosphorylation. CpdA, similarly to prednisolone, down-regulated endogenous and TNF-α-induced IL-6, IL-8, MMP-1 and MMP-3 protein secretion. The dissociative effect of CpdA was confirmed using chondrocytes with no induction of leptin secretion, but with a significant decrease in IL-6, IL-8, MMP-1 and MMP-3 protein secretion. CONCLUSION: CpdA, unlike prednisolone, did not induce leptin or Ob-R in human OA synovial fibroblasts, thereby demonstrating an improved risk:benefit ratio.


Subject(s)
Chondrocytes/metabolism , Fibroblasts/metabolism , Osteoarthritis/metabolism , Prednisolone/pharmacology , Receptors, Glucocorticoid/agonists , Synovial Membrane/metabolism , Aged , Aged, 80 and over , Blotting, Western , Chondrocytes/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Interleukin-6/metabolism , Interleukin-8/drug effects , Interleukin-8/metabolism , Leptin/metabolism , Male , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/metabolism , Middle Aged , Receptors, Leptin/drug effects , Receptors, Leptin/metabolism , Smad Proteins, Receptor-Regulated/drug effects , Smad Proteins, Receptor-Regulated/metabolism , Synovial Membrane/drug effects , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/metabolism , Tubulin/drug effects , Tubulin/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
12.
Scand J Rheumatol ; 44(3): 182-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25439190

ABSTRACT

OBJECTIVES: Progressive destruction of synovial joint cartilage and bone occurs in pathological conditions such as rheumatoid arthritis (RA) because of the overproduction of pro-inflammatory cytokines and activation of nuclear factor kappa B (NF-κB). Through the screening of NF-κB inhibitors by a luciferase reporter gene assay, we identified parthenolide (PAR) as the most potent NF-κB inhibitor, among several PAR analogue compounds. This study was undertaken to determine whether PAR inhibits pro-inflammatory cytokine production, cartilage degradation, and inflammatory arthritis. METHOD: The mRNA levels of pro-inflammatory cytokines were examined by real-time polymerase chain reaction (PCR). Proteoglycan content and release were determined by measuring glycosaminoglycan (GAG) levels using the dimethylmethylene blue (DMMB) dye-binding assay. The potential role of PAR in treatment of arthritis was studied using a collagen-induced arthritis (CIA) model. RESULTS: We established that PAR, as a prototype compound, suppressed lipopolysaccharide (LPS)- and tumour necrosis factor (TNF)-α-induced increases in matrix metalloproteinase (MMP)-1, MMP-3, inducible nitric oxide synthase (iNOS), and interleukin (IL)-1ß mRNA in chondrocytes. In addition, PAR prevented proteoglycan degradation triggered by pro-inflammatory cytokines. PAR treatment at the onset of CIA symptoms significantly reduced synovitis, inflammation, and pannus formation scores. Reduced synovial inflammation after PAR treatment was also reflected in significantly less bone erosion and cartilage damage. CONCLUSIONS: These data indicate a protective effect of PAR on the catabolic insults of pro-inflammatory cytokines on chondrocyte metabolism and GAG release in vitro and in CIA. PAR had anti-inflammatory and structure-modifying effects on experimental arthritis, suggesting that PAR may be useful as a potential alternative or adjunct therapy for inflammatory arthritis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Experimental , Cartilage, Articular/drug effects , Chondrocytes/drug effects , Cytokines/drug effects , NF-kappa B/antagonists & inhibitors , RNA, Messenger/drug effects , Sesquiterpenes/pharmacology , Synovial Membrane/drug effects , Animals , Cartilage, Articular/pathology , Chondrocytes/metabolism , Cytokines/metabolism , Disease Models, Animal , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/metabolism , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/metabolism , Rats , Synovial Membrane/pathology , Tumor Necrosis Factor-alpha/pharmacology
13.
Int J Cosmet Sci ; 37(2): 212-21, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25496720

ABSTRACT

OBJECTIVE: Quinoa (Chenopodium quinoa Willd.) is a seed crop rich in bioactive compounds including phytoecdysones (especially 20-hydroxyecdysone, 20HE), polyphenols, proteins and essential fatty acids. We previously reported a method to leach and concentrate quinoa bioactives into a complex phytochemical mixture termed quinoa leachate (QL). Here, we aimed to determine the effect of QL and its chemically distinct fractions on five biochemical endpoints relevant to skin care applications: (i) cell viability, (ii) matrix metalloproteinase (MMP) mRNA expression, (iii) MMP enzymatic activity, (iv) tyrosinase enzymatic activity and (v) intracellular reactive oxygen species (ROS) production. METHODS: Quinoa leachate was fractionated and chemically characterized using column chromatography and liquid chromatography-mass spectrometry (LC-MS). Cell viability was determined using a MTT assay in four mammalian cell lines. MMP-1 mRNA expression was assessed in human dermal fibroblasts (HDF) via qRT-PCR. The enzymatic activity of MMP-9 and tyrosinase was measured using fluorometric and colorimetric in vitro assays, respectively. Lipopolysaccharide (LPS)-induced ROS production was determined in human dermal fibroblasts by fluorescence intensity of an oxidant-sensitive probe. RESULTS: Quinoa leachate was separated into three fractions: (i) carbohydrate-rich fraction (QL-C; 71.3% w/w of QL); (ii) phytoecdysone, polyphenol and protein-rich fraction (QL-P, 13.3% w/w of QL); (iii) oil-rich fraction (QL-O, 10.8% w/w of QL). QL did not reduce cell viability in any of the four cell lines tested. QL, QL-P and QL-O each significantly inhibited MMP-1 mRNA expression in HDF at a concentration of 5 µg mL(-1) . QL and QL-P also significantly inhibited MMP-9 enzymatic activity, whereas QL-P demonstrated significant tyrosinase enzymatic inhibition. Furthermore, QL, QL-P, QL-O and 20HE significantly inhibited intracellular ROS production. CONCLUSION: This study is the first to demonstrate the MMP, tyrosinase and ROS inhibiting properties of multiple different phytochemical components derived from quinoa seeds. Our work indicates that quinoa phytochemicals may play a role in the treatment and prevention of skin ageing through a multiplicity of effects.


Subject(s)
Chenopodium quinoa/embryology , Matrix Metalloproteinase 1/drug effects , Protease Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Seeds/chemistry , Cells, Cultured , Chromatography, Liquid , Humans , Mass Spectrometry , Matrix Metalloproteinase 1/genetics , Monophenol Monooxygenase/antagonists & inhibitors
14.
J Cell Physiol ; 229(3): 292-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23929359

ABSTRACT

Interleukin-17 (IL-17) is a cytokine secreted predominantly by Th17 cells. Although IL-17 is primarily associated with the induction of tissue inflammation, the other biological functions of IL-17, including its wound-healing functions, have yet to be thoroughly explored. Fibroblast proliferation and migration play essential roles in periodontal wound-healing responses. In this study, we report that IL-17A can increase the migration and expression of matrix metalloproteinase (MMP)-1 in human periodontal ligament (PDL) fibroblasts but has no effect on PDL fibroblast proliferation. IL-17A-induced MMP-1 expression led to cell migration, which was attenuated by pre-treatment with IL-17 receptor neutralizing antibody and small interfering RNA (siRNA) for MMP-1. The IL-17A-induced cell migration was also attenuated by its tissue inhibitor of matrix metalloproteinase (TIMP)-1. In addition, a p38 mitogen-activated protein kinase (MAPK) inhibitor (SB203580) inhibited IL-17A-induced increase of the migration and MMP-1 upregulation of PDL fibroblasts. The involvement of p38 MAPK in IL-17A-induced MMP-1 expression and cell migration was further confirmed by transfection of p38α siRNA. A nuclear factor kappaB (NF-κB) inhibitor (pyrrolidine dithiocarbamate) also suppressed the cell migration and MMP-1 expression enhanced by IL-17A. Moreover, transfection with p38α siRNA inhibited IL-17A-induced NF-κB nuclear translocation as well as NF-κB binding activity. Our results suggest that IL-17A enhances the migration of PDL fibroblasts by increasing MMP-1 expression through the IL-17 receptor, p38 MAPK, and NF-κB signal transduction pathways.


Subject(s)
Cell Movement/drug effects , Fibroblasts/drug effects , Interleukin-17/pharmacology , Matrix Metalloproteinase 1/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , NF-kappa B/metabolism , Periodontal Ligament/drug effects , Active Transport, Cell Nucleus/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblasts/enzymology , Humans , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase Inhibitors/pharmacology , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/genetics , NF-kappa B/antagonists & inhibitors , Periodontal Ligament/cytology , Periodontal Ligament/enzymology , Protein Binding , Protein Kinase Inhibitors/pharmacology , RNA Interference , Signal Transduction/drug effects , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transfection
15.
Orthod Craniofac Res ; 17(1): 60-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-25125133

ABSTRACT

OBJECTIVES: To investigate the effects of IL-17 on IL-6, IL-1ß, and matrix metalloproteinase (MMP-1) production, and to compare the MMP-1 production between the individual and combined effects of IL-1ß and IL-6 in human periodontal ligament fibroblasts (HPDLF). MATERIALS AND METHODS: Human periodontal ligament fibroblasts were cultured with IL-17 for 0.5, 1, 4, 24, 48, and 72 h, and were cultured with IL-1ß, IL-6/sIL-6R, or a combination of IL-1ß and IL-6/sIL-6R for 24 h. To measure the mRNA levels of IL-6, IL-1ß, and MMP-1, total RNA was extracted from the cultured HPDLF, and a real-time PCR analysis was performed. The protein levels of IL-6, IL-1ß, and MMP-1 in supernatants were measured using enzyme-linked immunosorbent assays (ELISAs). RESULTS: IL-17 significantly increased the expression of IL-6 and MMP-1 mRNA and protein, while IL-17 transiently increased the expression of IL-1ß mRNA. The combination of IL-1ß and IL-6/sIL-6R induced significantly higher levels of MMP-1 protein than IL-1ß alone. CONCLUSIONS: IL-17 upregulated the production of IL-6 and MMP-1 sequentially in HPDLF. IL-6/sIL-6R may enhance the effects of IL-1ß on MMP-1 production. The present results suggest that IL-17 induces MMP-1 production not only directly, but also indirectly by promoting IL-6 production, thus resulting in the degradation of collagens in the PDL.


Subject(s)
Cytokines/drug effects , Fibroblasts/drug effects , Inflammation Mediators/analysis , Interleukin-17/pharmacology , Matrix Metalloproteinase 1/drug effects , Periodontal Ligament/drug effects , Cell Culture Techniques , Cells, Cultured , Fibroblasts/immunology , Humans , Interleukin-17/immunology , Interleukin-1beta/analysis , Interleukin-1beta/pharmacology , Interleukin-6/analysis , Interleukin-6/pharmacology , Matrix Metalloproteinase 1/analysis , Periodontal Ligament/cytology , Receptors, Interleukin-6/analysis , Receptors, Interleukin-6/immunology , Time Factors
16.
J Asian Nat Prod Res ; 16(12): 1139-47, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25465718

ABSTRACT

Chronic exposure to ultraviolet (UV) irradiation causes sunburn, inflammatory responses, skin cancer, and photoaging. Photoaging, in particular, generates reactive oxygen species (ROS) that stimulate mitogen-activated protein kinase (MAPK) signaling and transcription factors. UV irradiation also activates matrix metalloproteinases (MMPs) expression and inactivates collagen synthesis. Aaptamine, a marine alkaloid isolated from the marine sponge, has been reported to have antitumor, antimicrobial, antiviral, and antioxidant activities. However, the photo-protective effects of aaptamine have not been elucidated. In this study, our data demonstrated that aaptamine deactivated UVB-induced MAPK and activator protein-1 signaling by suppressing ROS, resulting in attenuating the expression of MMPs in UVB-irradiated human dermal fibroblasts. Aaptamine also decreased proinflammatory cytokines such as cyclooxygenase-2, tumor necrosis factor-α, interleukin-1ß, and nuclear factor-kappa B subunits in UVB-irradiated human keratinocytes. In conclusion, we suggest that aaptamine represents a novel and effective strategy for treatment and prevention of photoaging.


Subject(s)
Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 2/drug effects , Naphthyridines/pharmacology , Skin Diseases/prevention & control , Ultraviolet Rays/adverse effects , Epidermis/metabolism , Fibroblasts/drug effects , Humans , Interleukin-1beta/metabolism , Keratinocytes/drug effects , Molecular Structure , NF-kappa B/metabolism , Naphthyridines/chemistry , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Skin Aging/drug effects , Tumor Necrosis Factor-alpha/metabolism
17.
Acta Cir Bras ; 39: e391624, 2024.
Article in English | MEDLINE | ID: mdl-38808816

ABSTRACT

PURPOSE: To evaluate the chemotherapeutic activity of temozolomide counter to mammary carcinoma. METHODS: In-vitro anticancer activity has been conducted on MCF7 cells, and mammary carcinoma has been induced in Wistar rats by introduction of 7, 12-Dimethylbenz(a)anthracene (DMBA), which was sustained for 24 weeks. Histopathology, immunohistochemistry, cell proliferation study and apoptosis assay via TUNEL method was conducted to evaluate an antineoplastic activity of temozolomide in rat breast tissue. RESULTS: IC50 value of temozolomide in MCF7 cell has been obtained as 103 µM, which demonstrated an initiation of apoptosis. The temozolomide treatment facilitated cell cycle arrest in G2/M and S phase dose dependently. The treatment with temozolomide suggested decrease of the hyperplastic abrasions and renovation of the typical histological features of mammary tissue. Moreover, temozolomide therapy caused the downregulation of epidermal growth factor receptor, extracellular signal-regulated kinase, and metalloproteinase-1 expression and upstream of p53 and caspase-3 proliferation to indicate an initiation of apoptotic events. CONCLUSIONS: The occurrence of mammary carcinoma has been significantly decreased by activation of apoptotic pathway and abrogation of cellular propagation that allowable for developing a suitable mechanistic pathway of temozolomide in order to facilitate chemotherapeutic approach.


Subject(s)
Antineoplastic Agents, Alkylating , Apoptosis , ErbB Receptors , Rats, Wistar , Temozolomide , Temozolomide/pharmacology , Temozolomide/therapeutic use , Animals , Apoptosis/drug effects , Female , ErbB Receptors/drug effects , ErbB Receptors/antagonists & inhibitors , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/metabolism , Cell Proliferation/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Humans , MCF-7 Cells , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/drug effects , Immunohistochemistry , Reproducibility of Results , Rats , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology
18.
Am J Physiol Heart Circ Physiol ; 304(8): H1124-33, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23417861

ABSTRACT

We investigated the effect of cardiac-targeting erythropoietin (EPO)-encapsulated liposomes with sialyl Lewis(X) (SLX) on myocardial infarct (MI) size, left ventricular (LV) remodeling and function, and its molecular mechanism for repairing infarcted myocardium. In rabbits, MI was induced by 30 min of coronary occlusion followed by reperfusion. EPO-encapsulated liposomes with SLX (L-EPO group), EPO-encapsulated liposomes without SLX (L-EPO without SLX group), liposomes with SLX without EPO (L group), or saline (saline group) were intravenously administered immediately after MI. MI sizes and numbers of microvessels were assessed 14 days after MI. Prosurvival proteins and signals were assessed by Western blot analysis 2 and 14 days after MI. Confocal microscopy and electron microscopy showed the specific accumulation of liposomes with SLX in the infarcted myocardium. MI and cardiac fibrosis areas were significantly smaller in the L-EPO group than in the other groups. LV function and remodeling were improved in the L-EPO group. The number of CD31-positive microvessels was significantly greater in the L-EPO group than in the other groups. Higher expressions of EPO receptors, phosphorylated (p)Akt, pERK, pStat3, VEGF, Bcl-2, and promatrix metalloproteinase-1 were observed in the infarct area in the L-EPO group than in the other groups. EPO-encapsulated liposomes with SLX selectively accumulated in the infarct area, reduced MI size, and improved LV remodeling and function through activation of prosurvival signals and by exerting antifibrotic and angiogenic effects. EPO-encapsulated liposomes with SLX may be a promising strategy for active targeting treatment of acute MI.


Subject(s)
Erythropoietin/administration & dosage , Hematinics/administration & dosage , Myocardial Infarction/drug therapy , Ventricular Remodeling/drug effects , Animals , Drug Delivery Systems , Liposomes , Male , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Oligosaccharides/administration & dosage , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rabbits , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , Sialyl Lewis X Antigen , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology , Ventricular Remodeling/physiology , eIF-2 Kinase/drug effects , eIF-2 Kinase/metabolism
19.
Wound Repair Regen ; 21(1): 55-65, 2013.
Article in English | MEDLINE | ID: mdl-23126516

ABSTRACT

Here, we evaluate the efficacy of an emulgel dressing to control the release of an antifibrogenic factor, stratifin (SFN), along with an anti-inflammatory drug, acetylsalicylic acid (ASA), to be used as a wound dressing with hypertrophic scar reducing features. Emulgel dressings were prepared by dispersing positively charged submicron vesicles in carboxymethyl cellulose gel. Release kinetics of SFN/ASA and toxicity for primary skin cells were assessed in vitro. Antifibrogenic efficacy of medicated emulgel dressings was tested on a rabbit ear fibrotic model. Following topical application on the wounds, emulgels formed an occlusive film and controlled the release of SFN and ASA for 7 and 24 hours, respectively. Wounds treated with SFN/ASA-containing emulgel dressings showed an 80% reduction in scar elevation compared with untreated controls. Topical formulations were nontoxic for cultured human keratinocytes and fibroblasts. Inflammation was significantly controlled in treated wounds, as shown by a reduced number of infiltrated CD3(+) T cells (p < 0.001) and macrophages. SFN/ASA-treated wounds showed a significantly higher (p < 0.001) expression of matrix metalloproteinase-1, resulting in reduced collagen deposition and less scarring. Film-forming emulgel dressings that control the release of antifibrogenic and anti-inflammatory factors provide an excellent treatment option for postburn hypertrophic scar management.


Subject(s)
14-3-3 Proteins/pharmacology , Aspirin/pharmacology , Biomarkers, Tumor/pharmacology , Cicatrix, Hypertrophic/drug therapy , Delayed-Action Preparations/pharmacology , Exonucleases/pharmacology , Gels/pharmacology , Wound Healing , Wounds and Injuries/drug therapy , Administration, Cutaneous , Animals , Aspirin/administration & dosage , Bandages , Cicatrix, Hypertrophic/pathology , Cicatrix, Hypertrophic/prevention & control , Disease Models, Animal , Exoribonucleases , Fibroblasts/drug effects , Humans , Keratinocytes/drug effects , Matrix Metalloproteinase 1/drug effects , Rabbits , Wound Healing/drug effects , Wounds and Injuries/pathology
20.
J Periodontal Res ; 47(3): 320-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22092062

ABSTRACT

BACKGROUND AND OBJECTIVE: Marijuana (Cannabis sativa) use may be associated with gingival enlargement, resembling that caused by phenytoin. Cannabidiol (CBD), a nonpsychotropic Cannabis derivative, is structurally similar to phenytoin. While there are many reports on effects of phenytoin on human gingival fibroblasts, there is no information on effects of Cannabis components on these cells. The objective of this study was to determine effects of CBD on human gingival fibroblast fibrogenic and matrix-degrading activities. MATERIAL AND METHODS: Fibroblasts were incubated with CBD in serum-free medium for 1-6 d. The effect of CBD on cell viability was determined by measuring activity of a mitochondrial enzyme. The fibrogenic molecule transforming growth factor ß and the extracellular matrix molecule fibronectin were measured by ELISA. Pro-MMP-1 and total MMP-2 were measured by ELISA. Activity of MMP-2 was determined via a colorimetric assay in which a detection enzyme is activated by active MMP-2. Data were analysed using ANOVA and Scheffe's F procedure for post hoc comparisons. RESULTS: Cannabidiol had little or no significant effect on cell viability. Low CBD concentrations increased transforming growth factor ß production by as much as 40% (p < 0.001), while higher concentrations decreased it by as much as 40% (p < 0.0001). Cannabidiol increased fibronectin production by as much as approximately 100% (p < 0.001). Lower CBD concentrations increased MMP production, but the highest concentrations decreased production of both MMPs (p < 0.05) and decreased MMP-2 activity (p < 0.02). CONCLUSION: The data suggest that the CBD may promote fibrotic gingival enlargement by increasing gingival fibroblast production of transforming growth factor ß and fibronectin, while decreasing MMP production and activity.


Subject(s)
Cannabidiol/pharmacology , Extracellular Matrix/drug effects , Fibroblasts/drug effects , Fibronectins/drug effects , Gingiva/drug effects , Matrix Metalloproteinases/drug effects , Transforming Growth Factor beta/drug effects , Cannabidiol/toxicity , Cell Culture Techniques , Cell Survival/drug effects , Culture Media, Serum-Free , Enzyme Precursors/biosynthesis , Enzyme Precursors/drug effects , Extracellular Matrix/enzymology , Fibromatosis, Gingival/physiopathology , Fibronectins/biosynthesis , Gelatinases/biosynthesis , Gelatinases/drug effects , Gingiva/cytology , Humans , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/drug effects , Time Factors , Transforming Growth Factor beta/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL