Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters











Publication year range
1.
Mol Biol Rep ; 51(1): 608, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38704766

ABSTRACT

BACKGROUND: Tacrolimus (TAC) is a frequently used immunosuppressive medication in organ transplantation. However, its nephrotoxic impact limits its long-term usage. This study aims to investigate the effect of linagliptin (Lina) on TAC-induced renal injury and its underlying mechanisms. METHODS AND RESULTS: Thirty-two Sprague Dawley rats were treated with TAC (1.5 mg/kg/day, subcutaneously) and/or Lina (5 mg/kg/day, orally) for 4 weeks. Histological examination was conducted, and serum and urinary biomarkers were measured to assess kidney function and integrity. Furthermore, ELISA, Western blot analysis and immunohistochemical assay were employed to determine signaling molecules of oxidative stress, profibrogenic, hypoxic, and apoptotic proteins. Tacrolimus caused renal dysfunction and histological deterioration evidenced by increased serum creatinine, blood urea nitrogen (BUN), urinary cystatin C, and decreased serum albumin as well as elevated tubular injury and interstitial fibrosis scores. Additionally, TAC significantly increased the expression of collagen type-1, alpha-smooth muscle actin (α-SMA), plasminogen activator inhibitor-1 (PAI-1), and transforming growth factor-beta1 (TGF-ß1) renal content. Moreover, TAC decreased the expression of nuclear factor erythroid-2-related factor2 (Nrf2), heme oxygenase 1 (HO-1), and mitochondrial superoxide dismutase (SOD2). In addition, TAC increased protein expression of hypoxia-inducible factor1-alpha (HIF-1α), connective tissue growth factor (CTGF), inducible nitric oxide synthase (iNOS), 8-hydroxy-2-deoxyguanosine (8-OHdG), as well as nitric oxide (NO), 4-hydroxynonenal, caspase-3 and Bax renal contents. Furthermore, TAC decreased Bcl-2 renal contents. The Lina administration markedly attenuated these alterations. CONCLUSION: Lina ameliorated TAC-induced kidney injury through modulation of oxidative stress, hypoxia, and apoptosis related proteins.


Subject(s)
Acute Kidney Injury , Kidney , Linagliptin , NF-E2-Related Factor 2 , Oxidative Stress , Animals , Male , Rats , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/drug therapy , Connective Tissue Growth Factor/drug effects , Connective Tissue Growth Factor/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunosuppressive Agents/pharmacology , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Linagliptin/pharmacology , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Plasminogen Activator Inhibitor 1/drug effects , Plasminogen Activator Inhibitor 1/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tacrolimus/pharmacology , Tacrolimus/toxicity , Heme Oxygenase-1/drug effects , Heme Oxygenase-1/metabolism
2.
Biomed Pharmacother ; 145: 112402, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34773763

ABSTRACT

PAI-1 and CTGF are overexpressed in kidney diseases and cause fibrosis of the lungs, liver, and kidneys. We used a rat model of unilateral ureteral obstruction (UUO) to investigate whether 6-BIO, a glycogen synthase kinase-3ß inhibitor, attenuated fibrosis by inhibiting PAI-1 and CTGF in vivo. Additionally, TGFß-induced cellular fibrosis was observed in vitro using the human kidney proximal tubular epithelial cells (HK-2), and rat interstitial fibroblasts (NRK49F). Expression of fibrosis-related proteins and signaling molecules such as PAI-1, CTGF, TGFß, αSMA, SMAD, and MAPK were determined in HK-2 and NRK49F cells using immunoblotting. To identify the transcription factors that regulate the expression of PAI-1 and CTGF the promoter activities of AP-1 and SP-1 were analyzed using luciferase assays. Confocal microscopy was used to observe the co-localization of AP-1 and SP-1 to PAI-1 and CTGF. Expression of PAI-1, CTGF, TGFß, and α-SMA increased in UUO model as well as in TGFß-treated HK-2 and NRK49F cells. Furthermore, UUO and TGFß treatment induced the activation of P-SMAD2/3, SMAD4, P-ERK 1/2, P-P38, and P-JNK MAPK signaling pathways. PAI-1, CTGF, AP-1 and SP-1 promoter activity increased in response to TGFß treatment. However, treatment with 6-BIO decreased the expression of proteins and signaling pathways associated with fibrosis in UUO model as well as in TGFß-treated HK-2 and NRK49F cells. Moreover, 6-BIO treatment attenuated the expression of PAI-1 and CTGF as well as the promoter activities of AP-1 and SP-1, thereby regulating the SMAD and MAPK signaling pathways, and subsequently exerting anti-fibrotic effects on kidney cells.


Subject(s)
Indoles/pharmacology , Kidney Diseases/drug therapy , Kidney Tubules, Proximal/drug effects , Oximes/pharmacology , Animals , Cell Line , Connective Tissue Growth Factor/drug effects , Connective Tissue Growth Factor/genetics , Enzyme Inhibitors/pharmacology , Fibrosis , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Humans , Kidney Diseases/pathology , Kidney Tubules, Proximal/pathology , Male , Plasminogen Activator Inhibitor 1/drug effects , Plasminogen Activator Inhibitor 1/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sp1 Transcription Factor/drug effects , Sp1 Transcription Factor/genetics , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/genetics
3.
Am J Pathol ; 191(8): 1412-1430, 2021 08.
Article in English | MEDLINE | ID: mdl-34111429

ABSTRACT

Idiopathic subglottic stenosis (iSGS) is a progressive fibrotic disease characterized by life-threatening airway narrowing. Although the molecular underpinnings are unknown, previous reports showing that subglottic serial intralesional steroid injections (SILSIs) improve clinical outcomes suggest a steroid-sensitive pathway in iSGS. Herein, a prospective study was conducted to determine the changes in profibrotic markers during SILSI to identify steroid-sensitive profibrotic drivers. Seven newly diagnosed patients with iSGS were recruited for SILSI. Subglottic biopsies before and after SILSI treatments were evaluated for histologic and molecular markers by confocal microscopy and RT-qPCR. At baseline, iSGS subglottises contained abundant vimentin-positive/α-smooth muscle actin-negative fibroblasts, intermingled with a matrix of fibronectin and types I and VI collagen. Transforming growth factor (TGF)-ß1 was up-regulated primarily in glandular epithelium. Cellular communication network factor 2 (CCN2) was mainly up-regulated in stromal fibroblasts surrounding TGF-ß1-positive glandular structures. SILSI improved iSGS by reducing fibroblast infiltration and increasing matrix remodeling. Mechanistically, SILSI counteracted the effects of TGF-ß1 by inducing matrix metalloprotease 9 (MMP9) expression while repressing CCN2 expression, without affecting TGFß1 levels. Treatment of primary iSGS-derived fibroblasts with TGF-ß1 recapitulated aspects of the disease in vivo, demonstrating that the induction in CCN2 and repression of MMP9 are caused by changes in histone acetylation induced by TGF-ß1. Triamcinolone counteracted the coregulation of these genes by impairing SMAD2/3 binding to promoter regions, and not through histone acetylation. In conclusion, this study shows that SILSI counteracts a dysregulated TGF-ß1/CCN2/MMP9 axis involved in iSGS development.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Laryngostenosis/drug therapy , Signal Transduction/drug effects , Triamcinolone/therapeutic use , Connective Tissue Growth Factor/drug effects , Connective Tissue Growth Factor/metabolism , Down-Regulation , Humans , Injections, Intralesional , Laryngostenosis/metabolism , Laryngostenosis/pathology , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/metabolism , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/metabolism
4.
Wound Repair Regen ; 28(4): 460-469, 2020 07.
Article in English | MEDLINE | ID: mdl-32428986

ABSTRACT

Hypertrophic scar is an important clinical problem with limited therapeutic options. Aside from their roles as 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors, statins have also been demonstrated to decrease scarring by reducing connective tissue growth factor (CTGF) expression. However, poor penetrative ability limits their utility as topical treatments for hypertrophic scar. Here, we aim to develop novel statin formulations using liposomes to enhance dermal penetrative ability and to evaluate their efficacy against formation of hypertrophic scar utilizing our validated rabbit ear hypertrophic scar model. Liposomal simvastatin or pravastatin were compounded using a novel, flexible liposomal formulation and applied topically to rabbit ear hypertrophic scars daily from postoperation day (POD) 14 until POD 25. Scar color, including erythema and melanin, was measured using reflectance spectrophotometry on POD 28, and scar tissue was harvested for evaluation of scar elevation index as well as gene and protein expression. Human foreskin fibroblasts were also treated with statin formulations and CCN2 expression was determined by quantitative PCR. Both simvastatin and pravastatin were efficiently encapsulated in liposomes, forming nanometer-scale particles possessing highly negative charges. Topical treatment with liposomal simvastatin and pravastatin at 6.5% concentration significantly reduced scar elevation index and decreased type I/III collagen content and myofibroblast persistence in the wound. The erythema/vascularity of scars was reduced by liposomal statin treatment, with concomitant decrease of CD31 expression as measured histologically. Expression levels of transcripts encoding CTGF, collagen I, and collagen III collagen in scar tissue were also decreased by liposomal pravastatin treatment, as were myofibroblast persistence and the type I/III collagen ratio as assessed by immunofluorescence and picrosirus red staining, respectively. Treatment of human foreskin fibroblasts with simvastatin or with liposome-encapsulated pravastatin resulted in decreased expression of transcript encoding CTGF. Overall, our novel statin formulations encapsulated in liposomes were successfully delivered through topical application, significantly reducing hypertrophic scarring in a rabbit ear model.


Subject(s)
Cicatrix, Hypertrophic/metabolism , Fibroblasts/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Skin/metabolism , Animals , Cicatrix, Hypertrophic/pathology , Cicatrix, Hypertrophic/prevention & control , Collagen Type I/drug effects , Collagen Type I/genetics , Collagen Type III/drug effects , Collagen Type III/genetics , Connective Tissue Growth Factor/drug effects , Connective Tissue Growth Factor/genetics , Ear, External/injuries , Ear, External/metabolism , Ear, External/pathology , Erythema , Fibroblasts/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , In Vitro Techniques , Liposomes , Melanins , Platelet Endothelial Cell Adhesion Molecule-1/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pravastatin/administration & dosage , Pravastatin/pharmacology , Rabbits , Simvastatin/administration & dosage , Simvastatin/pharmacology , Skin/injuries , Skin/pathology , Spectrophotometry
6.
Lung ; 197(6): 783-791, 2019 12.
Article in English | MEDLINE | ID: mdl-31520180

ABSTRACT

BACKGROUND: Prolonged exposure to crystalline silica leads to persistent pulmonary inflammation and progressive fibrosis. Connective tissue growth factor (CTGF) has emerged as a potent proinflammatory and profibrotic regulator to participate in a variety of chronic inflammatory diseases. However, the role of CTGF in silica-induced pulmonary inflammation remains poorly understood. METHODS: To explore the effect of CTGF on inflammatory responses caused by silica particles, human bronchial epithelial cells (16HBE) were transfected with CTGF siRNA and exposed to silica particles at concentrations of 0, 12.5, 25, 50, 100 µg/ml for 48 h. Intracellular CTGF mRNA and protein expressions were determined by RT-PCR and Western blotting, respectively. The levels of inflammatory cytokines including IL-8, TNF-α, IL-6, IL-1ß, IL-17A and TGF-ß1 were measured by ELISA kits. RESULTS: Silica particles induce significantly elevated intracellular CTGF mRNA expression in 16HBE cells in a dose-dependent manner when compared with blank control group (P < 0.05). The secretions of IL-8, TNF-α, IL-6 and IL-17A were also significantly increased by silica particles (P < 0.05). After exposure to 25 or 50 µg/ml silica particles, the expression of intracellular CTGF mRNA was significantly inhibited in 16HBE cells when transfected with CTGF siRNA (P < 0.05). The secreted levels of IL-8, TNF-α, IL-6 and IL-17A induced by silica particles were also significantly lower from CTGF siRNA-transfected cells than that from normal 16HBE cells (P < 0.05). CONCLUSION: Inhibition of CTGF gene attenuates silica-induced inflammatory responses in bronchial epithelial cells, suggesting that CTGF could be a pivotal regulator in the development of silica-induced inflammation.


Subject(s)
Connective Tissue Growth Factor/drug effects , Epithelial Cells/drug effects , Inflammation/metabolism , Silicon Dioxide/pharmacology , Blotting, Western , Bronchi/cytology , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/immunology , Epithelial Cells/metabolism , Gene Knockdown Techniques , Humans , Inflammation/immunology , Interleukin-17/immunology , Interleukin-1beta/drug effects , Interleukin-1beta/immunology , Interleukin-6/immunology , Interleukin-8/drug effects , Interleukin-8/immunology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/cytology , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/immunology , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/immunology
7.
Can Respir J ; 2018: 8491487, 2018.
Article in English | MEDLINE | ID: mdl-30319721

ABSTRACT

Pulmonary fibrosis is a chronic and fatal disease of lung tissue with high incidence and mortality in the world. The exploration of effective treatment for pulmonary fibrosis remains an urgent challenge. In our study, Qingfei Xieding was investigated as a novel Chinese traditional patent medicine against pulmonary fibrosis. A pulmonary fibrosis mouse model was constructed by injecting with bleomycin sulfate. Following Qingfei Xieding administration, lung samples were collected to assess pulmonary phenotype changes by analyzing lung coefficient, wet/dry, and histopathologic section. Levels of nitric oxide (NO), hydroxyproline (HYP), malondialdehyde (MDA), and total antioxidant capacity were measured to evaluate the degree of oxidation. A single-cell gel electrophoresis (SCGE) assay was used to evaluate bleomycin-induced DNA damage. Western blotting and real-time quantitative PCR were performed to determine the abundance of inducible nitric oxide synthase (iNOS), connective tissue growth factor (CTGF), alpha smooth muscle actin (α-SMA), and fibronectin (FN). In the present study, Qingfei Xieding administration significantly attenuated bleomycin-induced pulmonary fibrosis in mice by reducing lung coefficient, wet/dry, NO, HYP, and MDA as well as the expression of iNOS, CTGF, α-SMA, FN, and DNA damage. The results indicated that Qingfei Xieding is effective to resist oxidative damage and histopathologic lesion, serving a protection role on bleomycin-induced pulmonary fibrosis.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Lung/drug effects , Pulmonary Fibrosis/metabolism , Actins/drug effects , Actins/genetics , Actins/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Bombyx , Connective Tissue Growth Factor/drug effects , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , DNA Damage/drug effects , Disease Models, Animal , Ephedra sinica , Fibronectins/drug effects , Fibronectins/genetics , Fibronectins/metabolism , Houttuynia , Hydroxyproline/drug effects , Hydroxyproline/metabolism , Lung/metabolism , Lung/pathology , Malondialdehyde/metabolism , Medicine, Chinese Traditional , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Prunus armeniaca , Pueraria , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Scutellaria baicalensis
8.
Rheumatology (Oxford) ; 57(9): 1675-1684, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29905853

ABSTRACT

Objectives: To investigate the mechanism of 2-methoxyestradiol (2-ME) in inhibiting hypoxia-induced collagen synthesis of fibroblasts in SSc. Methods: The expressions of hypoxia-inducible factor 1 alpha (HIF-1α) and connective tissue growth factor (CTGF) in skin specimens derived from SSc patients and healthy volunteers were examined by immunohistochemistry. HIF-1α was knocked down by lentiviral transduction, and SSc dermal fibroblasts cultured under normoxic (21% O2) or hypoxic (1% O2) condition were treated with PI3K inhibitor LY294002, rapamycin or 2-ME (25 µM). The protein levels of HIF-1α, CTGF, collagen I, p-Akt and p-mTOR were examined by western blotting or immunofluorescence. Apoptosis and cell cycle of fibroblasts were assessed by flow cytometry and by measuring caspase 3 activity, and cell proliferation was evaluated by Cell Counting Kit-8. Results: The expressions of HIF-1α and CTGF were increased in skins of SSc patients compared with healthy controls. Hypoxia up-regulated the protein levels of HIF-1α, CTGF and collagen I in SSc fibroblasts. In contrast, 2-ME inhibited PI3K/Akt/mTOR pathway and down-regulated protein levels of HIF-1α, CTGF and collagen I. Knockdown of HIF-1α reduced expressions of CTGF and collagen I, which were further down-regulated by 2-ME intervention. Moreover, 2-ME promoted the apoptosis and inhibited the proliferation of SSc fibroblasts by arresting the cell cycle at the G2/M phase. Conclusion: 2-ME reduced the production of CTGF and collagen I in SSc fibroblasts induced by hypoxia through PI3K/Akt/mTOR/HIF-1α signalling and inhibited the proliferation of fibroblasts. These findings suggested that 2-ME could be employed as a promising antifibrotic therapy for SSc.


Subject(s)
Collagen Type I/biosynthesis , Estradiol/analogs & derivatives , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Scleroderma, Systemic/genetics , TOR Serine-Threonine Kinases/genetics , 2-Methoxyestradiol , Cell Proliferation , Cells, Cultured , Collagen Type I/drug effects , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/drug effects , Estradiol/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/biosynthesis , RNA/genetics , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/metabolism , Signal Transduction , Skin/metabolism , Skin/pathology , TOR Serine-Threonine Kinases/biosynthesis , Tubulin Modulators/pharmacology
9.
Bratisl Lek Listy ; 118(8): 499-503, 2017.
Article in English | MEDLINE | ID: mdl-29050490

ABSTRACT

Vascular calcification is one of the most important factors for high morbidity and mortality from cardiovascular and cerebrovascular diseases. The aim of this study is to investigate the effect and mechanism of miR-26a on vascular smooth muscle cell calcification. First, the VSMCs were induced by ß-glycerol phosphate (ß-GP) for 7d and 14d, and Alizarin Red S staining was performed to examine the mineralized nodule change; then real time RT-PCR and western blotting were performed to explore the expression of miR-26a, CTGF, OPG, RANKL and ALP in un-induced and ß-GP-induced VSMCs; next, the VSMCs were transfected with miR-26a mimics, and Alizarin Red S staining was performed to examine the mineralized nodule change; finally, real time RT-PCR and western blotting were performed to explore the expression of miR-26a, CTGF, OPG, RANKL and ALP in un-transfected and miR-26a mimics transfected VSMCs. After ß-GP treatment, ß-GP promoted clear mineralized nodule changes, and miR-26a and OPG expression were significantly decreased and CTGF, RANKL and ALP expression were increased in VSMCs. Overexpression of miR-26a inhibited VSMCs calcification induced by ß-GP, and regulated the expression of CTGF, OPG, RANKL and ALP. Our findings suggested that up-regulation of miR-26a before ß-GP treatment inhibits VSMCs calcification through targeting CTGF (Fig. 4, Ref. 18).


Subject(s)
Connective Tissue Growth Factor/drug effects , Glycerophosphates/pharmacology , MicroRNAs/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Vascular Calcification/metabolism , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Cells, Cultured , Connective Tissue Growth Factor/metabolism , Humans , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Osteoprotegerin/drug effects , Osteoprotegerin/metabolism , RANK Ligand/drug effects , RANK Ligand/metabolism , Up-Regulation
10.
Endocrinology ; 158(10): 3620-3628, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28977597

ABSTRACT

Appropriate trophoblast invasion into the maternal endometrium is essential for successful human implantation and placentation. Connective tissue growth factor (CTGF), also known as CCN2, is a matricellular protein that is expressed in the placenta. Interestingly, the CTGF expression levels in the placenta and serum from patients with severe preeclampsia or fetal growth restriction are higher than those from healthy controls. However, to date, the role of CTGF in the regulation of trophoblast cell invasion remains unclear. Transforming growth factor-ß1 (TGF-ß1) is a potent stimulator of CTGF expression and has been shown to inhibit trophoblast cell invasiveness. However, whether CTGF mediates TGF-ß1-inhibited human trophoblast cell invasion is unknown. In the present study, we show that treatment with TGF-ß1 upregulates CTGF expression in a human trophoblast cell line, HTR-8/SVneo, and in primary human trophoblast cells. Our results also demonstrate that the SMAD2/3 signaling pathways are required for TGF-ß1-induced upregulation of CTGF. Importantly, CTGF knockdown attenuates TGF-ß1-inhibited cell invasion. Furthermore, cell invasiveness is decreased by treatment with recombinant CTGF. These results provide evidence that CTGF mediates TGF-ß1-inhibited human trophoblast cell invasion.


Subject(s)
Cell Movement/drug effects , Connective Tissue Growth Factor/drug effects , Transforming Growth Factor beta1/pharmacology , Trophoblasts/drug effects , Blotting, Western , Cell Line , Cell Movement/genetics , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Female , Gene Knockdown Techniques , Humans , Pregnancy , Pregnancy Trimester, First , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Trophoblasts/metabolism , Up-Regulation
11.
Burns ; 42(6): 1277-86, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27222384

ABSTRACT

Dermal wound healing, in which transforming growth factor beta 1 (TGFß1) plays an important role, is a complex process. Previous studies suggest that vitamin D has a potential regulatory role in TGFß1 induced activation in bone formation, and there is cross-talk between their signaling pathways, but research on their effects in other types of wound healing is limited. The authors therefore wanted to explore the role of vitamin D and its interaction with low concentration of TGFß1 in dermal fibroblast-mediated wound healing through an in vitro study. Human dermal fibroblasts were treated with vitamin D, TGFß1, both, or vehicle, and then the wound healing functions of dermal fibroblasts were measured. To further explore possible mechanisms explaining the synergistic effect of vitamin D and TGFß1, targeted gene silencing of the vitamin D receptor was performed. Compared to either factor alone, treatment of fibroblasts with both vitamin D and low concentration of TGFß1 increased gene expression of TGFß1, connective tissue growth factor, and fibronectin 1, and enhanced fibroblast migration, myofibroblast formation, and collagen production. Vitamin D receptor gene silencing blocked this synergistic effect of vitamin D and TGFß1 on both collagen production and myofibroblast differentiation. Thus a synergistic effect of vitamin D and low TGFß1 concentration was found in dermal fibroblast-mediated wound healing in vitro. This study suggests that supplementation of vitamin D may be an important step to improve wound healing and regeneration in patients with a vitamin D deficiency.


Subject(s)
Calcitriol/pharmacology , Dermis/drug effects , Fibroblasts/drug effects , Transforming Growth Factor beta1/pharmacology , Vitamins/pharmacology , Wound Healing/drug effects , Adult , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Chromatography, Liquid , Connective Tissue Growth Factor/drug effects , Connective Tissue Growth Factor/genetics , Dermis/metabolism , Drug Synergism , Female , Fibroblasts/metabolism , Fibronectins/drug effects , Fibronectins/genetics , Humans , Hydroxyproline/metabolism , In Vitro Techniques , Mass Spectrometry , Myofibroblasts/drug effects , Real-Time Polymerase Chain Reaction , Receptors, Calcitriol/drug effects , Receptors, Calcitriol/genetics , Reverse Transcriptase Polymerase Chain Reaction , Smad2 Protein/drug effects , Smad2 Protein/genetics , Smad3 Protein/drug effects , Smad3 Protein/genetics , Smad7 Protein/drug effects , Smad7 Protein/genetics , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/genetics
12.
Hum Exp Toxicol ; 35(2): 135-46, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25852135

ABSTRACT

Here we evaluated the ability of L-theanine in preventing experimental hepatic cirrhosis and investigated the roles of nuclear factor-κB (NF-κB) activation as well as transforming growth factor ß (TGF-ß) and connective tissue growth factor (CTGF) regulation. Experimental hepatic cirrhosis was established by the administration of carbon tetrachloride (CCl4) to rats (0.4 g/kg, intraperitoneally, three times per week, for 8 weeks), and at the same time, adding L-theanine (8.0 mg/kg) to the drinking water. Rats had ad libitum access to water and food throughout the treatment period. CCl4 treatment promoted NF-κB activation and increased the expression of both TGF-ß and CTGF. CCl4 increased the serum activities of alanine aminotransferase and γ-glutamyl transpeptidase and the degree of lipid peroxidation, and it also induced a decrease in the glutathione and glutathione disulfide ratio. L-Theanine prevented increased expression of NF-κB and down-regulated the pro-inflammatory (interleukin (IL)-1ß and IL-6) and profibrotic (TGF-ß and CTGF) cytokines. Furthermore, the levels of messenger RNA encoding these proteins decreased in agreement with the expression levels. L-Theanine promoted the expression of the anti-inflammatory cytokine IL-10 and the fibrolytic enzyme metalloproteinase-13. Liver hydroxyproline contents and histopathological analysis demonstrated the anti-fibrotic effect of l-theanine. In conclusion, L-theanine prevents CCl4-induced experimental hepatic cirrhosis in rats by blocking the main pro-inflammatory and pro-fibrogenic signals.


Subject(s)
Carbon Tetrachloride Poisoning/pathology , Carbon Tetrachloride Poisoning/prevention & control , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Connective Tissue Growth Factor/biosynthesis , Glutamates/therapeutic use , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , NF-kappa B/biosynthesis , Transforming Growth Factor beta/biosynthesis , Alanine Transaminase/blood , Animals , Antioxidants/metabolism , Aspartate Aminotransferases/blood , Connective Tissue Growth Factor/drug effects , Cytokines/biosynthesis , Down-Regulation/genetics , Lipid Peroxidation/drug effects , Liver Cirrhosis/chemically induced , Male , Matrix Metalloproteinase 13/biosynthesis , NF-kappa B/drug effects , Rats , Rats, Wistar , Transforming Growth Factor beta/drug effects
13.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 45(6): 598-606, 2016 05 25.
Article in Chinese | MEDLINE | ID: mdl-28247603

ABSTRACT

Objective: To investigate the role of glucose transporter 1 (GLUT1) and sodium-glucose cotransporter 1 (SGLT1) in high glucose dialysate-induced peritoneal fibrosis. Methods: Thirty six male SD rats were randomly divided into 6 groups (6 in each):normal control group, sham operation group, peritoneal dialysis group (PD group), PD+phloretin group (PD+T group), PD+phlorizin group (PD+Z group), PD+phloretin+phlorizin group (PD+T+Z group). Rat model of uraemia was established using 5/6 nephrotomy, and 2.5% dextrose peritoneal dialysis solution was used in peritoneal dialysis. Peritoneal equilibration test was performed 24 h after dialysis to evaluate transport function of peritoneum in rats; HE staining was used to observe the morphology of peritoneal tissue; and immunohistochemistry was used to detect the expression of GLUT1, SGLT1, TGF-ß1 and connective tissue growth factor (CTGF) in peritoneum. Human peritoneal microvascular endothelial cells (HPECs) were divided into 5 groups:normal control group, peritoneal dialysis group (PD group), PD+phloretin group (PD+T group), PD+phlorezin group (PD+Z group), and PD+phloretin+phlorezin group (PD+T+Z group). Real time PCR and Western blotting were used to detect mRNA and protein expressions of GLUT1, SGLT1, TGF-ß1, CTGF in peritoneal membrane and HPECs. Results:In vivo, compared with sham operation group, rats in PD group had thickened peritoneum, higher ultrafiltration volume, and the mRNA and protein expressions of GLUT1, SGLT1, CTGF, TGF-ß1 were significantly increased (all P<0.05); compared with PD group, thickened peritoneum was attenuated, and the mRNA and protein expressions of GLUT1, SGLT1, CTGF, TGF-ß1 were significantly decreased in PD+T, PD+Z and PD+T+Z groups (all P<0.05). Pearson's correlation analysis showed that the expressions of GLUT1, SGLT1 in peritoneum were positively correlated with the expressions of TGF-ß1 and CTGF (all P<0.05). In vitro, the mRNA and protein expressions of GLUT1, SGLT1, TGF-ß1, CTGF were significantly increased in HPECs of peritoneal dialysis group (all P<0.05), and those in PD+T, PD+Z, and PD+T+Z groups were decreased (all P<0.05). Pearson's correlation analysis showed that the expressions of GLUT1, SGLT1 in HPECs were positively correlated with the expressions of TGF-ß1 and CTGF (all P<0.05). Conclusion: High glucose peritoneal dialysis fluid may promote peritoneal fibrosis by upregulating the expressions of GLUT1 and SGLT1.


Subject(s)
Dialysis Solutions/adverse effects , Dialysis Solutions/pharmacology , Glucose Transporter Type 1/drug effects , Glucose Transporter Type 1/physiology , Glucose/adverse effects , Glucose/pharmacology , Hemodiafiltration/adverse effects , Hemodiafiltration/methods , Peritoneal Dialysis/adverse effects , Peritoneal Dialysis/methods , Peritoneal Fibrosis/chemically induced , Peritoneal Fibrosis/genetics , Peritoneum/chemistry , Peritoneum/drug effects , Peritoneum/pathology , Sodium-Glucose Transporter 1/drug effects , Sodium-Glucose Transporter 1/physiology , Uremia/chemically induced , Animals , Cells, Cultured , Connective Tissue Growth Factor/analysis , Connective Tissue Growth Factor/drug effects , Dialysis Solutions/chemistry , Gene Expression Regulation/drug effects , Glucose Transporter Type 1/analysis , Humans , Male , Peritoneal Fibrosis/physiopathology , Phloretin , Phlorhizin , RNA, Messenger , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 1/analysis , Transforming Growth Factor beta1/analysis , Transforming Growth Factor beta1/drug effects
14.
Oral Dis ; 21(6): 778-84, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25944709

ABSTRACT

OBJECTIVE: CCN family member 2/connective tissue growth factor (CCN2/CTGF) is known as an osteogenesis-related molecule and is thought to be implicated in tooth growth. Bone morphogenetic protein-1 (BMP-1) contributes to tooth development by the degradation of dentin-specific substrates as a metalloprotease. In this study, we demonstrated the correlations between CCN2/CTGF and BMP-1 in human carious teeth and the subcellular dynamics of BMP-1 in human dental pulp cells. MATERIALS AND METHODS: Expression of CCN2/CTGF and BMP-1 in human carious teeth was analyzed by immunohistochemistry. BMP-1-induced CCN2/CTGF protein expression in primary cultures of human dental pulp cells was observed by immunoblotting. Intracellular dynamics of exogenously administered fluorescence-labeled BMP-1 were observed using confocal microscope. RESULTS: Immunoreactivities for CCN2/CTGF and BMP-1 were increased in odontoblast-like cells and reparative dentin-subjacent dental caries. BMP-1 induced the expression of CCN2/CTGF independently of protease activity in the cells but not that of dentin sialophosphoprotein (DSPP) or dentin matrix protein-1 (DMP-1). Exogenously added BMP-1 was internalized into the cytoplasm, and the potent dynamin inhibitor dynasore clearly suppressed the BMP-1-induced CCN2/CTGF expression in the cells. CONCLUSION: CCN2/CTGF and BMP-1 coexist beneath caries lesion and CCN2/CTGF expression is regulated by dynamin-related cellular uptake of BMP-1, which suggests a novel property of metalloprotease in reparative dentinogenesis.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Connective Tissue Growth Factor/metabolism , Dental Pulp/metabolism , Dentinogenesis , Bone Morphogenetic Protein 1/analysis , Bone Morphogenetic Protein 1/pharmacology , Connective Tissue Growth Factor/analysis , Connective Tissue Growth Factor/drug effects , Dental Caries/metabolism , Dentin/chemistry , Extracellular Matrix Proteins/metabolism , Humans , Hydrazones/pharmacology , Phosphoproteins/metabolism , Primary Cell Culture , Sialoglycoproteins/metabolism , Young Adult
15.
J Periodontol ; 86(4): 569-77, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25594423

ABSTRACT

BACKGROUND: Enamel matrix derivative (EMD) is suggested to stimulate transforming growth factor-ß (TGF-ß) production. Connective tissue growth factor (CTGF) is a downstream mediator of TGF-ß. This study explores the effects of EMD and TGF-ß1 on CTGF in periodontal ligament (PDL) fibroblasts and their interactions in PDL proliferation and development. METHODS: Human PDL cells were stimulated with EMD. To explore the effects of EMD and TGF-ß1 on CTGF expression, cells were treated with and without TGF-ß inhibitor, and CTGF protein levels were assayed by Western blot analysis. To study the role of CTGF in PDL development, cells were treated with CTGF inhibitor. DNA synthesis was analyzed by bromodeoxyuridine enzyme-linked immunosorbent assay. Reverse-transcription polymerase chain reaction was performed to examine messenger RNA expression of PDL osteoblastic differentiation markers: type I collagen, alkaline phosphatase, and osteocalcin. RESULTS: EMD induced a concentration-dependent increase of CTGF protein expression in PDL cells. EMD- and TGF-ß1-stimulated CTGF expression was significantly reduced in the presence of TGF-ß inhibitor. CTGF inhibition downregulated both EMD- and TGF-ß1-induced DNA synthesis. The effect of CTGF and EMD on osteoblastic mRNA expression in PDL cells is not obvious. CONCLUSIONS: EMD stimulates CTGF expression in human PDL cells, a process modulated by the TGF-ß pathway. CTGF can affect EMD- and TGF-ß1-induced PDL cell proliferation, but its effects on PDL with regard to osteoblastic differentiation remain inconclusive. The results provide novel insights into EMD-CTGF interaction in PDL cells.


Subject(s)
Connective Tissue Growth Factor/drug effects , Dental Enamel Proteins/pharmacology , Fibroblasts/drug effects , Periodontal Ligament/drug effects , Transforming Growth Factor beta1/pharmacology , Adolescent , Adult , Alkaline Phosphatase/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/drug effects , Connective Tissue Growth Factor/antagonists & inhibitors , DNA/drug effects , Dental Enamel Proteins/administration & dosage , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Osteoblasts/drug effects , Osteocalcin/drug effects , Periodontal Ligament/cytology , RNA, Messenger/drug effects , Transforming Growth Factor beta1/antagonists & inhibitors , Young Adult
16.
J Oral Pathol Med ; 43(6): 464-70, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24484214

ABSTRACT

BACKGROUND: Oral submucous fibrosis (OSF) is a premalignant and fibrosing disease, which is closely associated with the habit of chewing areca nut. Panax notoginseng Buck F. H. Chen is an often used antifibrotic and antitumor agent. To treat areca nut-induced OSF, we have developed a chewable tablet, in which one of the major medicines is total Panax notoginseng saponins (PNS). In this study, we have investigated the antifibrotic effect and mechanism of PNS on areca nut-induced OSF in vitro. METHODS: Through human procollagen gene promoter luciferase reporter plasmid, hydroxyproline assay, gelatin zymography, qRT-PCR, ELISA, and Western blot, the influences of PNS on areca nut extract (ANE)-induced cell growth, collagen accumulation, procollagen gene transcription, MMP-2/-9 activity, MMP-1/-13 and TIMP-1/-2 expression, cytokine secretion, and the activation of PI3K/AKT, ERK/JNK/p38 MAPK, and TGFß/Smads pathways were detected. RESULTS: Panax notoginseng saponins could inhibit the ANE-induced abnormal growth and collagen accumulation of oral mucosal fibroblasts in a concentration-dependent manner. PNS (25 µg/ml) could significantly inhibit the ANE-induced expression of Col1A1 and Col3A1, augment the ANE-induced decrease of MMP-2/-9 activity, inhibit the ANE-induced increase of TIMP-1/-2 expression, and decrease the ANE-induced transcription and release of CTGF, TGFß1, IL-6, and TNFα. PNS (25 µg/ml) also significantly inhibited the ANE-induced activation of AKT and ERK/JNK/p38 MAPK pathways in oral mucosal fibroblasts and the ANE-induced activation of TGFß/smad pathway in HaCaT cells. CONCLUSION: Panax notoginseng saponins possess excellent anti-OSF activity, and its mechanism may be related to its ability to inhibit the ANE-induced activation of PI3K/AKT, ERK/JNK/p38 MAPK, and TGFß/smad pathways.


Subject(s)
Areca/adverse effects , Mouth Mucosa/drug effects , Nuts/adverse effects , Oral Submucous Fibrosis/pathology , Panax notoginseng , Plant Extracts/pharmacology , Saponins/pharmacology , Animals , Cell Culture Techniques , Cell Line , Collagen Type I/drug effects , Collagen Type I, alpha 1 Chain , Collagen Type III/drug effects , Connective Tissue Growth Factor/drug effects , Fibroblasts/drug effects , Humans , Hydroxyproline/analysis , Interleukin-6/analysis , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 2/drug effects , Matrix Metalloproteinase 9/drug effects , Mice , Mice, Inbred BALB C , Mouth Mucosa/cytology , Oral Submucous Fibrosis/etiology , Phosphatidylinositol 3-Kinases/drug effects , Plant Extracts/adverse effects , Proto-Oncogene Proteins c-akt/drug effects , Smad Proteins/drug effects , Tissue Inhibitor of Metalloproteinase-1/drug effects , Tissue Inhibitor of Metalloproteinase-2/drug effects , Transforming Growth Factor beta1/drug effects , Tumor Necrosis Factor-alpha/drug effects
17.
Pharm Biol ; 51(6): 712-21, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23373709

ABSTRACT

CONTEXT: Qindan capsule (QC), a compound used in traditional Chinese medicine, has been used as an anti-hypertensive agent in clinical settings for years. Our previous studies have shown that QC can improve the morphological index of the artery, down-regulate the collagen volume fraction in the media and inhibit the transformation of smooth muscle cells. However, the detailed mechanisms underlying its effects require further investigation, which might provide more scientific evidence for the clinical treatment of hypertensive vascular remodeling (VR). OBJECTIVE: We investigated the effects of QC-containing serum on the TGF-ß1/ERK signaling pathway, cell proliferation, migration, the cell cycle, apoptosis and matrix metalloproteinase synthesis (MMPs) in rat aortic adventitial fibroblasts (AFs). MATERIALS AND METHODS: AFs were cultured through tissue explants in vitro. The levels of extracellular signal-regulated kinase 1/2 (ERK1/2), phospho-ERK1/2 (p-ERK1/2), connective tissue growth factor (CTGF), MMP2 and MMP9 expression were measured by western blotting and RT-PCR. The proliferation and migration of AFs were measured by MTT and transwell migration assays. Cell cycle progression and apoptosis in AFs were analyzed by flow cytometry. RESULTS: The proliferation and migration rates of AFs treated with transforming growth factor ß1 (TGF-ß1) for 24 h were 2.4 ± 0.75 and 2.2 ± 0.06 times higher than those of untreated AFs, and increases in the expression of p-ERK1/2 (3.7 ± 0.15 times), CTGF (3.3 ± 0.24 times), MMP2 (5.7 ± 0.37 times) and MMP9 (5.4 ± 0.46 times) (p < 0.05) were observed. Treatment with QC-containing serum significantly down-regulated cell proliferation (1.9 ± 0.06 times), migration (1.6 ± 0.05 times) and the expression of p-ERK1/2 (1.3 ± 0.75 times), CTGF (1.8 ± 0.64 times), MMP2 (1.6 ± 0.65 times) and MMP9 (1.4 ± 0.46 times) (p < 0.05). We also found that QC-containing serum down-regulated the percentage of cells in the G1 phase by 1.6 ± 0.43 times and increased early-phase apoptosis by 2.3 ± 0.33 times (p < 0.05) in AFs. CONCLUSIONS: QC effectively inhibits the proliferation and migration of AFs and changes cell bioactivity and MMPs, possibly through the TGF-ß/ERK/CTGF signaling pathway. Our findings may provide new insights into the potential function of QC in preventing or treating hypertension.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Fibroblasts/drug effects , MAP Kinase Signaling System/drug effects , Transforming Growth Factor beta1/drug effects , Animals , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Connective Tissue Growth Factor/drug effects , Connective Tissue Growth Factor/metabolism , Drugs, Chinese Herbal/administration & dosage , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Medicine, Chinese Traditional , Rats , Rats, Inbred WKY , Serum/metabolism , Transforming Growth Factor beta1/metabolism
18.
J Periodontol ; 84(11): 1556-66, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23289869

ABSTRACT

BACKGROUND: Regeneration of periodontal tissues is one of the most important goals for the treatment of periodontal disease. The technology of plasma rich in growth factors provides a biologic approach for the stimulation and acceleration of tissue healing. The purpose of this study is to evaluate the biologic effects of this technology on primary human periodontal ligament fibroblasts. METHODS: The authors studied the response of periodontal ligament cells to this pool of growth factors on cell proliferation, cell migration, secretion of several biomolecules, cell adhesion, and expression of α2 integrin. Cell proliferation and adhesion were evaluated by means of a fluorescence-based method. Cell migration was performed on culture inserts. The release of different biomolecules by periodontal ligament fibroblasts was quantified through enzyme-linked immunosorbent assay. The α2 integrin expression was assessed through Western blot. RESULTS: This autologous technology significantly stimulated cell proliferation, migration, adhesion, and synthesis of many growth factors from cells including vascular endothelial growth factor, thrombospondin 1, connective tissue growth factor, hepatocyte growth factor, and procollagen type I. The α2 integrin expression was lower in plasma rich in growth factor-treated cells compared to non-stimulated cells, although no statistically significant differences were observed. CONCLUSION: This plasma rich in growth factors exerts positive effects on periodontal ligament fibroblasts, which could be positive for periodontal regeneration.


Subject(s)
Autografts/physiology , Periodontal Ligament/physiology , Platelet-Rich Plasma/physiology , Adolescent , Adult , Angiogenesis Inhibitors/analysis , Cell Adhesion/drug effects , Cell Culture Techniques , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen Type I/analysis , Collagen Type I/drug effects , Connective Tissue Growth Factor/analysis , Connective Tissue Growth Factor/drug effects , Endostatins/analysis , Female , Fibroblasts/drug effects , Fibroblasts/physiology , Hepatocyte Growth Factor/analysis , Humans , Insulin-Like Growth Factor I/analysis , Integrin alpha2/analysis , Integrin alpha2/drug effects , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Platelet-Derived Growth Factor/analysis , Platelet-Rich Plasma/chemistry , Regeneration/physiology , Thrombospondin 1/analysis , Thrombospondin 1/drug effects , Transforming Growth Factor beta1/analysis , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/drug effects , Young Adult
19.
Clin Oral Investig ; 17(2): 455-61, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22415218

ABSTRACT

OBJECTIVES: Transforming growth factor ß (TGFß) has been suggested as the main trigger for the increased collagen production and decreased matrix degradation pathways in oral submucous fibrosis (OSF). Connective tissue growth factor (CTGF/CCN2) and cyclooxygenase-2 (COX-2) were found to overexpress in OSF. The aim of this study was to investigate the molecular mechanism underlying the TGFß-induced CCN2 expressions in human buccal mucosal fibroblasts (BMFs) to identify the potential targets for drug intervention or chemoprevention of OSF. MATERIALS AND METHODS: TGFß-induced CCN2 expression and its signaling pathways were assessed by Western blot analyses in BMFs. RESULTS: TGFß1 stimulated CCN2 synthesis in BMFs. Pretreatment with c-Jun NH(2)-terminal kinase (JNK) inhibitor SP600125, p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580, and activin receptor-like kinase 5 (ALK5) inhibitor SB431542 significantly reduced TGFß1-induced CCN2 synthesis. Epigallocatechin-3-gallate (EGCG) completely blocked TGFß1-induced CCN2 synthesis by inhibiting the phosphorylation of JNK and p38 MAPK. Prostaglandin E(2) (PGE(2)) inhibited the TGFß1-induced CCN2 synthesis in human fetal lung fibroblasts IMR90 but not in BMFs. CONCLUSIONS: The TGFß1-induced CCN2 synthesis in BMFs could be mediated by the ALK5, JNK, and p38 MAPK pathways. EGCG blocks TGFß1-induced CCN2 by suppressing JNK and p38 in BMFs. CLINICAL RELEVANCE: The exceptional signal transduction pathways of TGFß1-induced CCN2 production in BMFs contribute to the resistance of PGE(2) downregulation of CCN2 expression; therefore, the CTGF/CCN2 levels are maintained in the OSF tissues in the presence of COX-2. EGCG may serve as a useful agent in controlling OSF.


Subject(s)
Catechin/analogs & derivatives , Connective Tissue Growth Factor/antagonists & inhibitors , Fibroblasts/drug effects , MAP Kinase Kinase 4/antagonists & inhibitors , Mouth Mucosa/drug effects , Protease Inhibitors/pharmacology , Transforming Growth Factor beta1/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Anthracenes/pharmacology , Benzamides/pharmacology , Catechin/pharmacology , Cell Line , Cells, Cultured , Connective Tissue Growth Factor/drug effects , Dinoprostone/pharmacology , Dioxoles/pharmacology , Dose-Response Relationship, Drug , Fibroblasts/enzymology , Flavonoids/pharmacology , Humans , Imidazoles/pharmacology , Lung/cytology , Lung/drug effects , Mouth Mucosa/cytology , Oral Submucous Fibrosis/enzymology , Oral Submucous Fibrosis/pathology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors
20.
Br J Ophthalmol ; 96(4): 587-90, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22289291

ABSTRACT

INTRODUCTION: In proliferative diabetic retinopathy (PDR), vascular endothelial growth factor (VEGF) and connective tissue growth factor (CTGF) may cause blindness by neovascularisation followed by fibrosis of the retina. It has previously been shown that a shift in the balance between levels of CTGF and VEGF in the eye is associated with this angiofibrotic switch. This study investigated whether anti-VEGF agents induce accelerated fibrosis in patients with PDR, as predicted by this model. METHODS: CTGF and VEGF levels were measured by ELISA in 52 vitreous samples of PDR patients, of which 24 patients had received intravitreal bevacizumab 1 week to 3 months before vitrectomy, and were correlated with the degree of vitreoretinal fibrosis as determined clinically and intra-operatively. RESULTS: CTGF correlated positively, and VEGF correlated negatively with the degree of fibrosis. The CTGF/VEGF ratio was the strongest predictor of fibrosis. Clinically, increased fibrosis was observed after intravitreal bevacizumab. CONCLUSIONS: These results confirm that the CTGF/VEGF ratio is a strong predictor of vitreoretinal fibrosis in PDR, and show that intravitreal anti-VEGF treatment causes increased fibrosis in PDR patients. These findings provide strong support for the model that the balance of CTGF and VEGF determines the angiofibrotic switch, and identify CTGF as a possible therapeutic target in the clinical management of PDR.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Connective Tissue Growth Factor/metabolism , Diabetic Retinopathy/metabolism , Retinal Neovascularization/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/administration & dosage , Bevacizumab , Connective Tissue Growth Factor/drug effects , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/surgery , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Humans , Intravitreal Injections , Male , Middle Aged , Retinal Neovascularization/drug therapy , Retinal Neovascularization/surgery , Vascular Endothelial Growth Factor A/drug effects , Vitrectomy , Vitreous Body/metabolism , Vitreous Body/surgery
SELECTION OF CITATIONS
SEARCH DETAIL