1.Osthole decreases collagen I/III contents and their ratio in TGF-β1-overexpressed mouse cardiac fibroblasts through regulating the TGF-β/Smad signaling pathway.
Jin-Cheng LIU ; Lei ZHOU ; Feng WANG ; Zong-Qi CHENG ; Chen RONG
Chinese Journal of Natural Medicines (English Ed.) 2018;16(5):321-329
		                        		
		                        			
		                        			The present study was designed to elucidate whether the mechanism by which osthole decreases collagenI/III contents and their ratio is regulating the TGF-β/Smad signaling pathway in TGF-β1-overexpressed mouse cardiac fibroblasts (CFs). These CFs were cultured and treated with different concentrations of osthole. Our results showed that the TGF-β1 expression in the CFs transfected with that the recombinant expression plasmids pcDNA3.1(+)-TGF-β1 was significantly enhanced. After the CFs were treated with 1.25-5 μg·mL of osthole for 24 h, the mRNA and protein expression levels of collagensIand III were reduced. The collagen I/III ratio was also reduced. The mRNA and protein expression levels of TGF-β1, TβRI, Smad2/3, P-Smad2/3, Smad4, and α-SMA were decreased, whereas the expression level of Smad7 was increased. These effects suggested that osthole could inhibit collagen I and III expression and reduce their ratio via the TGF-β/Smad signaling pathway in TGF-β1 overexpressed CFs. These effects of osthole may play beneficial roles in the prevention and treatment of myocardial fibrosis.
		                        		
		                        		
		                        		
		                        			Actins
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Cells, Cultured
		                        			;
		                        		
		                        			Collagen
		                        			;
		                        		
		                        			biosynthesis
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Coumarins
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Fibroblasts
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Gene Expression Regulation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Myocardium
		                        			;
		                        		
		                        			cytology
		                        			;
		                        		
		                        			Protein-Serine-Threonine Kinases
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			RNA, Messenger
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Real-Time Polymerase Chain Reaction
		                        			;
		                        		
		                        			Receptor, Transforming Growth Factor-beta Type I
		                        			;
		                        		
		                        			Receptors, Transforming Growth Factor beta
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Signal Transduction
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Smad Proteins
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Transforming Growth Factor beta1
		                        			;
		                        		
		                        			genetics
		                        			
		                        		
		                        	
2.Aldosterone induces inflammatory cytokines in penile corpus cavernosum by activating the NF-κB pathway.
Fei WU ; Zu-Quan XIONG ; Shan-Hua MAO ; Ji-Meng HU ; Jian-Qing WANG ; Hao-Wen JIANG ; Qiang DING
Asian Journal of Andrology 2018;20(1):24-29
		                        		
		                        			
		                        			Emerging evidence indicates that aldosterone and mineralocorticoid receptors (MRs) are associated with the pathogenesis of erectile dysfunction. However, the molecular mechanisms remain largely unknown. In this study, freshly isolated penile corpus cavernosum tissue from rats was treated with aldosterone, with or without MRs inhibitors. Nuclear factor (NF)-kappa B (NF-κB) activity was evaluated by real-time quantitative PCR, luciferase assay, and immunoblot. The results demonstrated that mRNA levels of the NF-κB target genes, including inhibitor of NF-κB alpha (IκB-α), NF-κB1, tumor necrosis factor-alpha (TNF-α), and interleukin 6 (IL-6), were higher after aldosterone treatment. Accordingly, phosphorylation of p65/RelA, IκB-α, and inhibitor of NF-κB kinase-β was markedly increased by aldosterone. Furthermore, knockdown of MRs prevented activation of the NF-κB canonical pathway by aldosterone. Consistent with this finding, ectopic overexpression of MRs enhanced the transcriptional activation of NF-κB by aldosterone. More importantly, the MRs antagonist, spironolactone blocked aldosterone-mediated activation of the canonical NF-κB pathway. In conclusion, aldosterone has an inflammatory effect in the corpus cavernosum penis, inducing NF-κB activation via an MRs-dependent pathway, which may be prevented by selective MRs antagonists. These data reveal the possible role of aldosterone in erectile dysfunction as well as its potential as a novel pharmacologic target for treatment.
		                        		
		                        		
		                        		
		                        			Aldosterone/pharmacology*
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Cytokines/biosynthesis*
		                        			;
		                        		
		                        			Gene Knockdown Techniques
		                        			;
		                        		
		                        			I-kappa B Kinase/antagonists & inhibitors*
		                        			;
		                        		
		                        			Interleukin-6/genetics*
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Mineralocorticoid Receptor Antagonists/pharmacology*
		                        			;
		                        		
		                        			NF-kappa B/genetics*
		                        			;
		                        		
		                        			Penis/metabolism*
		                        			;
		                        		
		                        			Protein Serine-Threonine Kinases/antagonists & inhibitors*
		                        			;
		                        		
		                        			RNA, Messenger/biosynthesis*
		                        			;
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Rats, Inbred WKY
		                        			;
		                        		
		                        			Receptors, Mineralocorticoid/genetics*
		                        			;
		                        		
		                        			Signal Transduction/drug effects*
		                        			;
		                        		
		                        			Spironolactone/pharmacology*
		                        			;
		                        		
		                        			Transcriptional Activation
		                        			;
		                        		
		                        			Tumor Necrosis Factor-alpha/biosynthesis*
		                        			;
		                        		
		                        			NF-kappaB-Inducing Kinase
		                        			
		                        		
		                        	
3.Effects of estrogen receptor GPR30 agonist G1 on neuronal apoptosis and microglia polarization in traumatic brain injury rats.
Meng-Xian PAN ; Jun-Chun TANG ; Rui LIU ; Yu-Gong FENG ; Qi WAN
Chinese Journal of Traumatology 2018;21(4):224-228
PURPOSETo investigate the effects of estrogen G protein-coupled receptor 30 (GPR30) agonist G1 on hippocampal neuronal apoptosis and microglial polarization in rat traumatic brain injury (TBI).
METHODSMale SD rats were randomly divided into sham group, TBI + vehicle group, TBI + G1 group. Experimental moderate TBI was induced using Feeney's weigh-drop method. G1 (100μg/kg) or vehicle was intravenously injected from femoral vein at 30 min post-injury. Rats were sacrificed at 24 h after injury for detection of neuronal apoptosis and microglia polarization. Neuronal apoptosis was assayed by immunofluorescent staining of active caspase-3. M1 type microglia markers (iNOS and IL-1β) and M2 type markers (Arg1 and IL-4) were examined by immunoblotting or ELISA. Total protein level of Akt and phosphorylated Akt were assayed by immunoblotting.
RESULTSG1 significantly reduced active caspase-3 positive neurons in hippocampus. Meanwhile G1 increased the ratio of Arg1/iNOS. IL-1β production was decreased but IL-4 was increased after G1 treatment. G1 treatment also increased the active form of Akt.
CONCLUSIONSGPR30 agonist G1 inhibited neuronal apoptosis and favored microglia polarization to M2 type.
Animals ; Apoptosis ; drug effects ; Brain Injuries, Traumatic ; drug therapy ; pathology ; Cell Polarity ; Hippocampus ; drug effects ; Interleukin-1beta ; biosynthesis ; Male ; Microglia ; drug effects ; Neurons ; drug effects ; Proto-Oncogene Proteins c-akt ; metabolism ; Rats ; Rats, Sprague-Dawley ; Receptors, G-Protein-Coupled ; agonists
4.IFN-γ secretion in gut of Ob/Ob mice after vertical sleeve gastrectomy and its function in weight loss mechanism.
Jin-Peng DU ; Geng WANG ; Chao-Jie HU ; Qing-Bo WANG ; Hui-Qing LI ; Wen-Fang XIA ; Xiao-Ming SHUAI ; Kai-Xiong TAO ; Guo-Bin WANG ; Ze-Feng XIA
Journal of Huazhong University of Science and Technology (Medical Sciences) 2016;36(3):377-382
		                        		
		                        			
		                        			Vertical sleeve gastrectomy (VSG) is becoming more and more popular among the world. Despite its dramatic efficacy, however, the mechanism of VSG remains largely undetermined. This study aimed to test interferon (IFN)-γ secretion n of mesenteric lymph nodes in obese mice (ob/ob mice), a model of VSG, and its relationship with farnesoid X receptor (FXR) expression in the liver and small intestine, and to investigate the weight loss mechanism of VSG. The wild type (WT) mice and ob/ob mice were divided into four groups: A (WT+Sham), B (WT+VSG), C (ob/ob+Sham), and D (ob/ob+VSG). Body weight values were monitored. The IFN-γ expression in mesenteric lymph nodes of ob/ob mice pre- and post-operation was detected by flow cytometry (FCM). The FXR expression in the liver and small intestine was detected by Western blotting. The mouse AML-12 liver cells were stimulated with IFN-γ at different concentrations in vitro. The changes of FXR expression were also examined. The results showed that the body weight of ob/ob mice was significantly declined from (40.6±2.7) g to (27.5±3.8) g on the 30th day after VSG (P<0.05). At the same time, VSG induced a higher level secretion of IFN-γ in mesenteric lymph nodes of ob/ob mice than that pre-operation (P<0.05). The FXR expression levels in the liver and small intestine after VSG were respectively 0.97±0.07 and 0.84±0.07 fold of GAPDH, which were significantly higher than pre-operative levels of 0.50±0.06 and 0.48±0.06 respectively (P<0.05). After the stimulation of AML-12 liver cells in vitro by different concentrations of IFN-γ (0, 10, 25, 50, 100, and 200 ng/mL), the relative FXR expression levels were 0.22±0.04, 0.31±0.04, 0.39±0.05, 0.38±0.05, 0.56±0.06, and 0.35±0.05, respectively, suggesting IFN-γ could distinctly promote the FXR expression in a dose-dependent manner in comparison to those cells without IFN-γ stimulation (P<0.05). It was concluded that VSG induces a weight loss in ob/ob mice by increasing IFN-γ secretion of mesenteric lymph nodes, which then increases the FXR expression of the liver and small intestine.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Body Weight
		                        			;
		                        		
		                        			Cell Line
		                        			;
		                        		
		                        			Gastrectomy
		                        			;
		                        		
		                        			methods
		                        			;
		                        		
		                        			Gene Expression
		                        			;
		                        		
		                        			Hepatocytes
		                        			;
		                        		
		                        			cytology
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Interferon-gamma
		                        			;
		                        		
		                        			biosynthesis
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			secretion
		                        			;
		                        		
		                        			Intestine, Small
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Liver
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Lymph Nodes
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Mesentery
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Obese
		                        			;
		                        		
		                        			Obesity
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			surgery
		                        			;
		                        		
		                        			Receptors, Cytoplasmic and Nuclear
		                        			;
		                        		
		                        			agonists
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Weight Loss
		                        			
		                        		
		                        	
5.Effect of curcumin on aged Drosophila melanogaster: a pathway prediction analysis.
Zhi-guo ZHANG ; Xu-yan NIU ; Ai-ping LU ; Gary Guishan XIAO
Chinese journal of integrative medicine 2015;21(2):115-122
OBJECTIVETo re-analyze the data published in order to explore plausible biological pathways that can be used to explain the anti-aging effect of curcumin.
METHODSMicroarray data generated from other study aiming to investigate effect of curcumin on extending lifespan of Drosophila melanogaster were further used for pathway prediction analysis. The differentially expressed genes were identified by using GeneSpring GX with a criterion of 3.0-fold change. Two Cytoscape plugins including BisoGenet and molecular complex detection (MCODE) were used to establish the protein-protein interaction (PPI) network based upon differential genes in order to detect highly connected regions. The function annotation clustering tool of Database for Annotation, Visualization and Integrated Discovery (DAVID) was used for pathway analysis.
RESULTSA total of 87 genes expressed differentially in D. melanogaster melanogaster treated with curcumin were identified, among which 50 were up-regulated significantly and 37 were remarkably down-regulated in D. melanogaster melanogaster treated with curcumin. Based upon these differential genes, PPI network was constructed with 1,082 nodes and 2,412 edges. Five highly connected regions in PPI networks were detected by MCODE algorithm, suggesting anti-aging effect of curcumin may be underlined through five different pathways including Notch signaling pathway, basal transcription factors, cell cycle regulation, ribosome, Wnt signaling pathway, and p53 pathway.
CONCLUSIONGenes and their associated pathways in D. melanogaster melanogaster treated with anti-aging agent curcumin were identified using PPI network and MCODE algorithm, suggesting that curcumin may be developed as an alternative therapeutic medicine for treating aging-associated diseases.
Aging ; drug effects ; genetics ; Animals ; Cell Cycle ; drug effects ; genetics ; Curcumin ; pharmacology ; Drosophila Proteins ; genetics ; metabolism ; Drosophila melanogaster ; drug effects ; genetics ; Gene Expression Regulation ; drug effects ; Gene Regulatory Networks ; drug effects ; Genes, Insect ; Protein Biosynthesis ; drug effects ; genetics ; Protein Interaction Maps ; drug effects ; genetics ; Receptors, Notch ; genetics ; metabolism ; Ribosomes ; drug effects ; metabolism ; Signal Transduction ; drug effects ; genetics ; Tumor Suppressor Protein p53 ; metabolism ; Wnt Signaling Pathway ; drug effects ; genetics
6.In silico target fishing for the potential bioactive components contained in Huanglian Jiedu Tang (HLJDD) and elucidating molecular mechanisms for the treatment of sepsis.
Shi-Tang MA ; Cheng-Tao FENG ; Guo-Liang DAI ; Yue SONG ; Guo-Liang ZHOU ; Xiao-Lin ZHANG ; Cheng-Gui MIAO ; Hao YU ; Wen-Zheng JU
Chinese Journal of Natural Medicines (English Ed.) 2015;13(1):30-40
		                        		
		                        			
		                        			The present study was designed to target fish for potential bioactive components contained in a Huang Lian Jie Du decoction (HLJDD) and identify the underlying mechanisms of action for the treatment of sepsis at the molecular level. he bioactive components database of HLJDD was constructed and the sepsis-associated targets were comprehensively investigated. The 3D structures of the PAFR and TXA2R proteins were established using the homology modelling (HM) method, and the molecular effects for sepsis treatment were analysed by comparing the bioactive components database and the sepsis targets using computational biology methods. The results of the screening were validated with biological testing against the human oral epidermal carcinoma cell line KB in vitro. We found that multiple bioactive compounds contained in the HLJDD interacted with multiple targets. We also predicted the promising compound leads for sepsis treatment, and the first 28 compounds were characterized. Several compounds, such as berberine, berberrubine and epiberberine, dose-dependently inhibited PGE2 production in human KB cells, and the effects were similar in the presence or absence of TPA. This study demonstrates a novel approach to identifying natural chemical compounds as new leads for the treatment of sepsis.
		                        		
		                        		
		                        		
		                        			Anti-Inflammatory Agents, Non-Steroidal
		                        			;
		                        		
		                        			pharmacokinetics
		                        			;
		                        		
		                        			Berberine
		                        			;
		                        		
		                        			analogs & derivatives
		                        			;
		                        		
		                        			pharmacokinetics
		                        			;
		                        		
		                        			Dinoprostone
		                        			;
		                        		
		                        			biosynthesis
		                        			;
		                        		
		                        			Drugs, Chinese Herbal
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			pharmacokinetics
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			KB Cells
		                        			;
		                        		
		                        			Platelet Membrane Glycoproteins
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Protein Transport
		                        			;
		                        		
		                        			Receptors, G-Protein-Coupled
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Receptors, Thromboxane A2, Prostaglandin H2
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Sepsis
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Tetradecanoylphorbol Acetate
		                        			;
		                        		
		                        			pharmacokinetics
		                        			
		                        		
		                        	
7.Wheat peptides reduce oxidative stress and inhibit NO production through modulating μ-opioid receptor in a rat NSAID-induced stomach damage model.
Hong YIN ; Hui-Zhen CAI ; Shao-Kang WANG ; Li-Gang YANG ; Gui-Ju SUN
Chinese Journal of Natural Medicines (English Ed.) 2015;13(1):22-29
		                        		
		                        			
		                        			Non-steroidal anti-inflammatory drugs (NSAIDs) induce tissue damage and oxidative stress in animal models of stomach damage. In the present study, the protective effects of wheat peptides were evaluated in a NSAID-induced stomach damage model in rats. Different doses of wheat peptides or distilled water were administered daily by gavage for 30 days before the rat stomach damage model was established by administration of NSAIDs (aspirin and indomethacin) into the digestive tract twice. The treatment of wheat peptides decreased the NSAID-induced gastric epithelial cell degeneration and oxidative stress and NO levels in the rats. Wheat peptides significantly increased the superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities and decreased iNOS activity in stomach. The mRNA expression level of μ-opioid receptor was significantly decreased in wheat peptides-treated rats than that in in the control rats. The results suggest that NSAID drugs induced stomach damage in rats, wchih can be prevented by wheat peptides. The mechanisms for the protective effects were most likely through reducing NSAID-induced oxidative stress.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Anti-Inflammatory Agents, Non-Steroidal
		                        			;
		                        		
		                        			adverse effects
		                        			;
		                        		
		                        			Antioxidants
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Aspirin
		                        			;
		                        		
		                        			adverse effects
		                        			;
		                        		
		                        			Gastric Mucosa
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Gene Expression
		                        			;
		                        		
		                        			Glutathione Peroxidase
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Indomethacin
		                        			;
		                        		
		                        			adverse effects
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Nitric Oxide
		                        			;
		                        		
		                        			biosynthesis
		                        			;
		                        		
		                        			Nitric Oxide Synthase
		                        			;
		                        		
		                        			chemical synthesis
		                        			;
		                        		
		                        			Oxidation-Reduction
		                        			;
		                        		
		                        			Oxidative Stress
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Plant Proteins
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			RNA, Messenger
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Rats, Sprague-Dawley
		                        			;
		                        		
		                        			Receptors, Opioid, mu
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Stomach
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Superoxide Dismutase
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Triticum
		                        			;
		                        		
		                        			chemistry
		                        			
		                        		
		                        	
8.Glucocorticoid receptor agonist dexamethasone attenuates renal ischemia/reperfusion injury by up-regulating eNOS/iNOS.
Jiong ZHANG ; Jun-hua LI ; Le WANG ; Min HAN ; Fang XIAO ; Xiao-qin LAN ; Yue-qiang LI ; Gang XU ; Ying YAO
Journal of Huazhong University of Science and Technology (Medical Sciences) 2014;34(4):516-520
		                        		
		                        			
		                        			The aim of this study was to determine the effect of dexamethasone (DEX) on renal ischemia/reperfusion injury (IRI). C57BL/6 mice were randomly divided into Sham group, IRI group and DEX group. The mice in IRI and DEX groups subjected to renal ischemia for 60 min, were treated with saline or DEX (4 mg/kg, i.p.) 60 min prior to I/R. After 24 h of reperfusion, the renal function, renal pathological changes, activation of extracellular signal-regulated kinase (ERK) and glucocorticoid receptor (GR), and the levels of iNOS and eNOS were detected. The results showed DEX significantly decreased the damage to renal function and pathological changes after renal IRI. Pre-treatment with DEX reduced ERK activation and down-regulated the level of iNOS, whereas up-regulated the level of eNOS after renal IRI. DEX could further promote the activation of GR. These findings indicated GR activation confers preconditioning-like protection against acute IRI partially by up-regulating the ratio of eNOS/iNOS.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Dexamethasone
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Gene Expression Regulation, Enzymologic
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Glucocorticoids
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Nitric Oxide Synthase Type II
		                        			;
		                        		
		                        			biosynthesis
		                        			;
		                        		
		                        			Nitric Oxide Synthase Type III
		                        			;
		                        		
		                        			biosynthesis
		                        			;
		                        		
		                        			Receptors, Glucocorticoid
		                        			;
		                        		
		                        			agonists
		                        			;
		                        		
		                        			Reperfusion Injury
		                        			;
		                        		
		                        			enzymology
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Up-Regulation
		                        			;
		                        		
		                        			drug effects
		                        			
		                        		
		                        	
9.Influences of IL-6R antibody on PMMA bone cement-mediated expression of OPG and RANKL in synovial fibroblasts.
Ke TAO ; Hui ZENG ; De-ming XIAO ; Ao XIONG ; Jian WENG ; Bin KANG
Journal of Huazhong University of Science and Technology (Medical Sciences) 2014;34(2):241-246
		                        		
		                        			
		                        			Effect of interleukin-6 receptor (IL-6R) antibody on polymethyl methacrylate (PMMA) bone cement-mediated expression of osteoprotegerin (OPG) and receptor activator of nuclear factor-kappaB ligand (RANKL) in synovial fibroblasts was investigated. Synovial tissue obtained from total knee arthroplasty was digested and cultured. Inverted microscope was employed to observe the synovial cells and immunocytochemistry (SABC method) staining was used to identify synovial fibroblasts. This experiment was divided into three groups according to different culture media: PMMA group (75 μg/mL PMMA bone cement particles), IL-6R antibody group (10 ng/mL IL-6R antibody+75 μg/mL PMMA bone cement particles), and control group (no IL-6R antibody or PMMA bone cement particles). Influence of IL-6R antibody and PMMA on proliferation of synovial fibroblasts was measured by cell counting kit-8 (CCK-8). ELISA method was used to measure OPG and RANKL levels in culture solution. Fluorescence quantitative real-time PCR (FQ-PCR) was used to detect the expression of OPG and RANKL mRNA. After three consecutive passages, more than 95% of the primary synovial cells became long spindle fibroblast-like cells. SABC staining results showed that the fibroblast-like cells were negative for anti-CD68 antibody and positive for anti-vimentin antibody, with brown madder stained. CCK-8 test demonstrated that the absorbance (A) value at 450 nm was significantly lower in IL-6R antibody group than in PMMA group and control group (P<0.01), but there was no statistically significant difference in A value at 450 nm between the control group and PMMA group (P>0.05). Results of ELISA indicated that the expression of OPG was significantly higher in IL-6R antibody group than in PMMA group and control group (P<0.01). The expression of RANKL was inhibited (P<0.05), and the ratio of OPG/RANKL was significantly increased in IL-6R antibody group as compared with PMMA group and control group. There was no significant difference in the expression of OPG between control group and PMMA group (P>0.05), but the expression of RANKL was higher in PMMA group than in control group (P<0.05), and there was a significant difference in the ratio of OPG/RANKL between them (P<0.05). Results of FQ-PCR revealed the expression of RANKL mRNA was significantly inhibited (P<0.01) and the expression of OPG mRNA was significantly increased (P<0.01) in IL-6R antibody group as compared with PMMA group and control group. The expression of RANKL mRNA was higher in PMMA group than in control group (P<0.05), but the expression of OPG mRNA had no significant difference between them (P>0.05). IL-6R antibody could significantly increase the expression of OPG, but inhibit the expression of RANKL, which might provide a theoretical basis of molecular biology for the prevention and treatment of aseptic loosening of prosthesis.
		                        		
		                        		
		                        		
		                        			Antibodies
		                        			;
		                        		
		                        			administration & dosage
		                        			;
		                        		
		                        			immunology
		                        			;
		                        		
		                        			Bone Cements
		                        			;
		                        		
		                        			Fibroblasts
		                        			;
		                        		
		                        			immunology
		                        			;
		                        		
		                        			Gene Expression
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Osteoprotegerin
		                        			;
		                        		
		                        			biosynthesis
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Polymethyl Methacrylate
		                        			;
		                        		
		                        			administration & dosage
		                        			;
		                        		
		                        			Prostheses and Implants
		                        			;
		                        		
		                        			RANK Ligand
		                        			;
		                        		
		                        			biosynthesis
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Receptors, Interleukin-6
		                        			;
		                        		
		                        			immunology
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Synovial Fluid
		                        			;
		                        		
		                        			immunology
		                        			;
		                        		
		                        			metabolism
		                        			
		                        		
		                        	
10.Effect of Bilirubin on Triglyceride Synthesis in Streptozotocin-Induced Diabetic Nephropathy.
Jianwei XU ; Eun Seong LEE ; Seon Ha BAEK ; Shin Young AHN ; Sejoong KIM ; Ki Young NA ; Dong Wan CHAE ; Ho Jun CHIN
Journal of Korean Medical Science 2014;29(Suppl 2):S155-S163
		                        		
		                        			
		                        			We aimed to elucidate the effect of bilirubin on dyslipidemia and nephropathy in a diabetes mellitus (DM) type I animal model. Sprague-Dawley rats were separated into control, DM, and bilirubin-treated DM (Bil) groups. The Bil group was injected intraperitoneally with 60 mg/kg bilirubin 3 times per week and hepatoma cells were cultured with bilirubin at a concentration of 0.3 mg/dL. The Bil group showed lower serum creatinine levels 5 weeks after diabetes onset. Bilirubin treatment also decreased the amount of mesangial matrix, lowered the expression of renal collagen IV and transforming growth factor (TGF)-beta1, and reduced the level of apoptosis in the kidney, compared to the DM group. These changes were accompanied by decreased tissue levels of hydrogen superoxide and NADPH oxidase subunit proteins. Bilirubin decreased serum total cholesterol, high-density lipoprotein cholesterol (HDL-C), free fatty acids, and triglycerides (TGs), as well as the TG content in the liver tissues. Bilirubin suppressed protein expression of LXRalpha, SREBP-1, SCD-1, and FAS, factors involved in TG synthesis that were elevated in the livers of DM rats and hepatoma cells under high-glucose conditions. In conclusion, bilirubin attenuates renal dysfunction and dyslipidemia in diabetes by suppressing LXRalpha and SREBP-1 expression and oxidative stress.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Bilirubin/pharmacology/*therapeutic use
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Creatine/blood
		                        			;
		                        		
		                        			Diabetes Mellitus, Experimental/chemically induced/complications/*pathology
		                        			;
		                        		
		                        			Diabetic Nephropathies/*drug therapy/etiology
		                        			;
		                        		
		                        			Disease Models, Animal
		                        			;
		                        		
		                        			Kidney/pathology
		                        			;
		                        		
		                        			Lipoproteins, HDL/blood
		                        			;
		                        		
		                        			Liver/metabolism
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Inbred C57BL
		                        			;
		                        		
		                        			NADPH Oxidase/metabolism
		                        			;
		                        		
		                        			Orphan Nuclear Receptors/antagonists & inhibitors/genetics/metabolism
		                        			;
		                        		
		                        			Oxidative Stress/drug effects
		                        			;
		                        		
		                        			Rats
		                        			;
		                        		
		                        			Rats, Sprague-Dawley
		                        			;
		                        		
		                        			Reactive Oxygen Species/metabolism
		                        			;
		                        		
		                        			Streptozocin/toxicity
		                        			;
		                        		
		                        			Triglycerides/analysis/*biosynthesis/blood
		                        			
		                        		
		                        	
            
Result Analysis
Print
Save
E-mail