1.Effect of RAD18-siRNA on proliferation and chemotherapy sensitivity of human esophageal squamous cell carcinoma ECA-109 cells.
Pengrong LOU ; Xiaonan SUN ; Jundong ZHOU ; Shitao ZOU
Journal of Zhejiang University. Medical sciences 2016;45(4):364-370
To investigate the effect of RAD18-siRNA on cell proliferation and chemotherapy sensitivity of esophageal squamous cell carcinoma (ESCC) ECA-109 cells.RAD18-siRNA was transfected into human ECA-109 cells by Lipofectamine 3000. Quantitative PCR and Western blot were performed to detect RAD18 and CyclinD1 expression; CCK-8 assay was used to determine cell proliferation and chemotherapy drug sensitivity; flow cytometry was used to determine cell cycle. Correlation between RAD18 and CyclinD1 mRNA expression was analyzed by Pearson's correlation.Compared with non-transfected cells, the expression of RAD18 in RAD18-siRNA group was significantly decreased (<0.05). The cell proliferation was inhibited (<0.05) and the cell number of G1 phase was increased, G2/M phase cells decreased (<0.05) in RAD18-siRNA group. After treatment with different concentrations of cisplatin or 5-FU, the survival rate of the two cell groups was reduced (all<0.05), and the IC50 of RAD18-siRNA group was significantly lower than that of non-transfected group (<0.05). The mRNA expression of RAD18 was positively correlated with CyclinD1 expression in ESCC tissues(=0.478,<0.01).Down-regulated expression of RAD18 can decrease the cell proliferation and increase chemo-sensitivity of ESCC cells, and CyclinD1 may participate in the process.
Adjuvants, Pharmaceutic
;
pharmacology
;
Carcinoma, Squamous Cell
;
drug therapy
;
physiopathology
;
Cell Cycle
;
Cell Line, Tumor
;
Cell Proliferation
;
drug effects
;
Cisplatin
;
pharmacology
;
Cyclin D1
;
drug effects
;
genetics
;
DNA-Binding Proteins
;
administration & dosage
;
pharmacology
;
Down-Regulation
;
drug effects
;
genetics
;
Drug Resistance, Neoplasm
;
drug effects
;
Drug Screening Assays, Antitumor
;
methods
;
Drug Synergism
;
Esophageal Neoplasms
;
drug therapy
;
physiopathology
;
Fluorouracil
;
pharmacology
;
G1 Phase
;
drug effects
;
G2 Phase
;
drug effects
;
Humans
;
Metaphase
;
drug effects
;
RNA, Small Interfering
;
administration & dosage
;
pharmacology
;
Transfection
;
Ubiquitin-Protein Ligases
;
administration & dosage
;
pharmacology
2.Use of deferasirox, an iron chelator, to overcome imatinib resistance of chronic myeloid leukemia cells.
Dae Sik KIM ; Yoo Jin NA ; Myoung Hee KANG ; Soo Young YOON ; Chul Won CHOI
The Korean Journal of Internal Medicine 2016;31(2):357-366
BACKGROUND/AIMS: The treatment of chronic myeloid leukemia (CML) has achieved impressive success since the development of the Bcr-Abl tyrosine kinase inhibitor, imatinib mesylate. Nevertheless, resistance to imatinib has been observed, and a substantial number of patients need alternative treatment strategies. METHODS: We have evaluated the effects of deferasirox, an orally active iron chelator, and imatinib on K562 and KU812 human CML cell lines. Imatinib-resistant CML cell lines were created by exposing cells to gradually increasing concentrations of imatinib. RESULTS: Co-treatment of cells with deferasirox and imatinib induced a synergistic dose-dependent inhibition of proliferation of both CML cell lines. Cell cycle analysis showed an accumulation of cells in the subG1 phase. Western blot analysis of apoptotic proteins showed that co-treatment with deferasirox and imatinib induced an increased expression of apoptotic proteins. These tendencies were clearly identified in imatinib-resistant CML cell lines. The results also showed that co-treatment with deferasirox and imatinib reduced the expression of BcrAbl, phosphorylated Bcr-Abl, nuclear factor-kappaB (NF-kappaB) and beta-catenin. CONCLUSIONS: We observed synergistic effects of deferasirox and imatinib on both imatinib-resistant and imatinib-sensitive cell lines. These effects were due to induction of apoptosis and cell cycle arrest by down-regulated expression of NF-kappaB and beta-catenin levels. Based on these results, we suggest that a combination treatment of deferasirox and imatinib could be considered as an alternative treatment option for imatinib-resistant CML.
Antineoplastic Agents/*pharmacology
;
Apoptosis/drug effects
;
Apoptosis Regulatory Proteins/metabolism
;
Benzoates/*pharmacology
;
Cell Proliferation/drug effects
;
Dose-Response Relationship, Drug
;
Drug Resistance, Neoplasm/*drug effects
;
G1 Phase Cell Cycle Checkpoints/drug effects
;
Humans
;
Imatinib Mesylate/*pharmacology
;
Iron Chelating Agents/*pharmacology
;
K562 Cells
;
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/*drug therapy/metabolism
;
Protein Kinase Inhibitors/*pharmacology
;
Signal Transduction/drug effects
;
Triazoles/*pharmacology
3.Novel matrine derivative MD-1 attenuates hepatic fibrosis by inhibiting EGFR activation of hepatic stellate cells.
Yi FENG ; Hai-Yan YING ; Ying QU ; Xiao-Bo CAI ; Ming-Yi XU ; Lun-Gen LU
Protein & Cell 2016;7(9):662-672
Matrine (MT), the effective component of Sophora flavescens Ait, has been shown to have anti-inflammation, immune-suppressive, anti-tumor, and anti-hepatic fibrosis activities. However, the pharmacological effects of MT still need to be strengthened due to its relatively low efficacy and short half-life. In the present study, we report a more effective thio derivative of MT, MD-1, and its inhibitory effects on the activation of hepatic stellate cells (HSCs) in both cell culture and animal models. Cytological experiments showed that MD-1 can inhibit the proliferation of HSC-T6 cells with a half-maximal inhibitory concentration (IC50) of 62 μmol/L. In addition, MD-1 more strongly inhibits the migration of HSC-T6 cells compared to MT and can more effectively induce G0/G1 arrest and apoptosis. Investigating the biological mechanisms underlying anti-hepatic fibrosis in the presence of MD-1, we found that MD-1 can bind the epidermal growth factor receptor (EGFR) on the surface of HSC-T6 cells, which can further inhibit the phosphorylation of EGFR and its downstream protein kinase B (Akt), resulting in decreased expression of cyclin D1 and eventual inhibition of the activation of HSC-T6 cells. Furthermore, in rats with dimethylnitrosamine (DMN)-induced hepatic fibrosis, MD-1 slowed the development and progression of hepatic fibrosis, protecting hepatic parenchymal cells and improving hepatic functions. Therefore, MD-1 is a potential drug for anti-hepatic fibrosis.
Alkaloids
;
pharmacology
;
Animals
;
Cell Line
;
Cyclin D1
;
metabolism
;
Dimethylnitrosamine
;
toxicity
;
Enzyme Activation
;
drug effects
;
ErbB Receptors
;
metabolism
;
G1 Phase Cell Cycle Checkpoints
;
drug effects
;
Hepatic Stellate Cells
;
metabolism
;
pathology
;
Liver Cirrhosis
;
chemically induced
;
metabolism
;
pathology
;
prevention & control
;
Phosphorylation
;
drug effects
;
Proto-Oncogene Proteins c-akt
;
metabolism
;
Quinolizines
;
pharmacology
;
Rats
4.Total triterpenoids from Ganoderma Lucidum suppresses prostate cancer cell growth by inducing growth arrest and apoptosis.
Tao WANG ; Zi-ping XIE ; Zhan-sen HUANG ; Hao LI ; An-yang WEI ; Jin-ming DI ; Heng-jun XIAO ; Zhi-gang ZHANG ; Liu-hong CAI ; Xin TAO ; Tao QI ; Di-ling CHEN ; Jun CHEN
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(5):736-741
In this study, one immortalized human normal prostatic epithelial cell line (BPH) and four human prostate cancer cell lines (LNCaP, 22Rv1, PC-3, and DU-145) were treated with Ganoderma Lucidum triterpenoids (GLT) at different doses and for different time periods. Cell viability, apoptosis, and cell cycle were analyzed using flow cytometry and chemical assays. Gene expression and binding to DNA were assessed using real-time PCR and Western blotting. It was found that GLT dose-dependently inhibited prostate cancer cell growth through induction of apoptosis and cell cycle arrest at G1 phase. GLT-induced apoptosis was due to activation of Caspases-9 and -3 and turning on the downstream apoptotic events. GLT-induced cell cycle arrest (mainly G1 arrest) was due to up-regulation of p21 expression at the early time and down-regulation of cyclin-dependent kinase 4 (CDK4) and E2F1 expression at the late time. These findings demonstrate that GLT suppresses prostate cancer cell growth by inducing growth arrest and apoptosis, which might suggest that GLT or Ganoderma Lucidum could be used as a potential therapeutic drug for prostate cancer.
Antineoplastic Agents, Phytogenic
;
isolation & purification
;
pharmacology
;
Apoptosis
;
drug effects
;
Caspase 3
;
genetics
;
metabolism
;
Caspase 9
;
genetics
;
metabolism
;
Cell Line, Tumor
;
Cell Survival
;
drug effects
;
Cyclin D1
;
genetics
;
metabolism
;
Cyclin-Dependent Kinase 4
;
genetics
;
metabolism
;
Cyclin-Dependent Kinase Inhibitor p21
;
genetics
;
metabolism
;
Dose-Response Relationship, Drug
;
E2F1 Transcription Factor
;
genetics
;
metabolism
;
G1 Phase Cell Cycle Checkpoints
;
drug effects
;
genetics
;
Gene Expression Regulation, Neoplastic
;
Humans
;
Male
;
Nucleosomes
;
drug effects
;
metabolism
;
pathology
;
Plant Extracts
;
chemistry
;
Prostate
;
drug effects
;
metabolism
;
pathology
;
Reishi
;
chemistry
;
Signal Transduction
;
Triterpenes
;
isolation & purification
;
pharmacology
5.A novel schiff base zinc coordination compound inhibits proliferation and induces apoptosis of human osteosarcoma cells.
Ming YAN ; Li PANG ; Tan-tan MA ; Cheng-liang ZHAO ; Nan ZHANG ; Bing-xin YU ; Yan XIA
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(5):700-706
Various kinds of schiff base metal complexes have been proven to induce apoptosis of tumor cells. However, it remains largely unknown whether schiff base zinc complexes induce apoptosis in human cancer cells. Here, we synthesized a novel schiff base zinc coordination compound (SBZCC) and investigated its effects on the growth, proliferation and apoptosis of human osteosarcoma MG-63 cells. A novel SBZCC was synthesized by chemical processes and used to treat MG-63 cells. The cell viability was determined by CCK-8 assay. The cell cycle progression, mitochondrial membrane potential and apoptotic cells were analyzed by flow cytometry. The apoptosis-related proteins levels were determined by immunoblotting. Treatment of MG-63 cells with SBZCC resulted in inhibition of cell proliferation and cell cycle arrest at G1 phase. Moreover, SBZCC significantly reduced the mitochondrial membrane potential and induced apoptosis, accompanied with increased Bax/Bcl-2 and FlasL/Fas expression as well as caspase-3/8/9 cleavage. Our results demonstrated that the synthesized novel SBZCC could inhibit the proliferation and induce apoptosis of MG-63 cells via activating both the mitochondrial and cell death receptor apoptosis pathways, suggesting that SBZCC is a promising agent for the development as anticancer drugs.
Antineoplastic Agents
;
chemical synthesis
;
pharmacology
;
Apoptosis
;
drug effects
;
Caspase 3
;
genetics
;
metabolism
;
Caspase 8
;
genetics
;
metabolism
;
Caspase 9
;
genetics
;
metabolism
;
Cell Line, Tumor
;
Cell Proliferation
;
drug effects
;
Cell Survival
;
drug effects
;
Coordination Complexes
;
chemical synthesis
;
pharmacology
;
Fas Ligand Protein
;
genetics
;
metabolism
;
G1 Phase Cell Cycle Checkpoints
;
drug effects
;
Gene Expression Regulation, Neoplastic
;
drug effects
;
Humans
;
Membrane Potential, Mitochondrial
;
drug effects
;
Mitochondria
;
drug effects
;
metabolism
;
pathology
;
Osteoblasts
;
drug effects
;
metabolism
;
pathology
;
Proto-Oncogene Proteins c-bcl-2
;
genetics
;
metabolism
;
Schiff Bases
;
chemistry
;
Signal Transduction
;
Zinc
;
chemistry
;
bcl-2-Associated X Protein
;
genetics
;
metabolism
;
fas Receptor
;
genetics
;
metabolism
6.Kinetin inhibits proliferation of hepatic stellate cells by interrupting cell cycle and induces apoptosis by down-regulating ratio of Bcl-2/Bax.
Zhen-gang ZHANG ; Jie ZOU ; Ying HUANG ; Liang WU
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(5):672-678
Liver fibrosis is an important health problem that can further progress into cirrhosis or liver cancer, and result in significant morbidity and mortality. Inhibiting proliferation and inducing apoptosis of hepatic stellate cells (HSCs) may be the key point to reverse liver fibrosis. At present, anti-fibrosis drugs are rare. Kinetin is a type of plant-derived cytokinin which has been reported to control differentiation and induce apoptosis of human cells. In this study, the HSCs were incubated with different concentrations of kinetin. The proliferation of rat HSCs was measured by MTT assay, cell cycle and apoptosis were analyzed by flow cytometry, and the apoptosis was examined by TUNEL method. The expression of Bcl-2 and Bax proteins was detected by immunocytochemistry staining. It was found that kinetin could markedly inhibit proliferation of HSCs. In a concentration range of 2 to 8 μg/mL, the inhibitory effects of kinetin on proliferation of HSCs were increased with the increased concentration and the extension of time (P < 0.01). Flow cytometry indicated that kinetin could inhibit the DNA synthesis from G0/G1 to S phase in a dose-dependent manner (P < 0.01). The apoptosis rates of the HSCs treated with 8, 4 and 2 μg/mL kinetin (25.62% ± 2.21%, 15.31% ± 1.9% and 6.18% ± 1.23%, respectively) were increased significantly compared with the control group (3.81% ± 0.93%) (P < 0.01). All the DNA frequency histogram in kinetin-treated groups showed obvious hypodiploid peak (sub-G1 peak), and with the increase of kinetin concentrations, the apoptosis rate of HSCs also showed a trend of increase. It was also found that kinetin could down-regulate the expression of Bcl-2, and up-regulate the expression of Bax, leading to the decreased ratio of Bcl-2/Bax significantly. The kinetin-induced apoptosis of HSCs was positively correlated with the expression of Bax, and negatively with the expression of Bcl-2. It was concluded that kinetin can inhibit activation and proliferation of HSCs by interrupting the cell cycle at G1/S restriction point and inducing apoptosis of HSCs via reducing the ratio of Bcl-2/Bax.
Animals
;
Apoptosis
;
drug effects
;
Cell Line, Transformed
;
Cell Proliferation
;
drug effects
;
Dose-Response Relationship, Drug
;
G1 Phase Cell Cycle Checkpoints
;
drug effects
;
genetics
;
Gene Expression Regulation
;
Growth Inhibitors
;
pharmacology
;
Hepatic Stellate Cells
;
cytology
;
drug effects
;
metabolism
;
Kinetin
;
pharmacology
;
Proto-Oncogene Proteins c-bcl-2
;
antagonists & inhibitors
;
genetics
;
metabolism
;
Rats
;
Signal Transduction
;
bcl-2-Associated X Protein
;
agonists
;
genetics
;
metabolism
7.Influence of P53 on the radiotherapy response of hepatocellular carcinoma.
Ana R GOMES ; Ana M ABRANTES ; Ana F BRITO ; Mafalda LARANJO ; Joao E CASALTA-LOPES ; Ana C GONCALVES ; Ana B SARMENTO-RIBEIRO ; Maria F BOTELHO ; Jose G TRALHAO
Clinical and Molecular Hepatology 2015;21(3):257-267
BACKGROUND/AIMS: Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide, and it has a poor prognosis and few therapeutic options. Radiotherapy is one of the most effective forms of cancer treatment, and P53 protein is one of the key molecules determining how a cell responds to radiotherapy. The aim of this study was to determine the therapeutic efficacy of iodine-131 in three human HCC cell lines. METHODS: Western blotting was used to measure P53 expression. The effects of radiotherapy with iodine-131 were assessed by using the clonogenic assay to evaluate cell survival. Flow cytometry was carried out to examine the effects of iodine-131 on cell death, oxidative stress, reduced intracellular glutathione expression, the mitochondrial membrane potential, and the cell cycle. RESULTS: The P53 protein was not expressed in Hep3B2.1-7 cells, was expressed at normal levels in HepG2 cells, and was overexpressed in HuH7 cells. P53 expression in the HuH7 and HepG2 cell lines increased after internal and external irradiation with iodine-131. Irradiation induced a decrease in cell survival and led to a decrease in cell viability in all of the cell lines studied, accompanied by cell death via late apoptosis/necrosis and necrosis. Irradiation with 131-iodine induced mostly cell-cycle arrest in the G0/G1 phase. CONCLUSIONS: These results suggest that P53 plays a key role in the radiotherapy response of HCC.
Apoptosis/*radiation effects
;
Blotting, Western
;
Carcinoma, Hepatocellular/metabolism/pathology/radiotherapy
;
Cell Line, Tumor
;
Cell Survival/drug effects
;
G1 Phase Cell Cycle Checkpoints/radiation effects
;
*Gamma Rays
;
Glutathione/metabolism
;
Hep G2 Cells
;
Humans
;
Iodine Radioisotopes/chemistry/pharmacology/therapeutic use
;
Liver Neoplasms/metabolism/pathology/radiotherapy
;
Phosphorylation
;
Reactive Oxygen Species/metabolism
;
Tumor Suppressor Protein p53/*metabolism
8.Marsdenia tenacissima extract induces G0/G1 cell cycle arrest in human esophageal carcinoma cells by inhibiting mitogen-activated protein kinase (MAPK) signaling pathway.
Wei FAN ; Li SUN ; Jing-Qian ZHOU ; Cang ZHANG ; Song QIN ; Ying TANG ; Yang LIU ; Sen-Sen LIN ; Sheng-Tao YUAN
Chinese Journal of Natural Medicines (English Ed.) 2015;13(6):428-437
Marsdenia tenacissima extract (MTE, trade name: Xiao-Ai-Ping injection) is an extract of a single Chinese plant medicine. It has been used for the treatment of cancer in China for decades, especially for esophageal cancer and other cancers in the digestive tract. In the present study, the potential mechanism for MTE's activity in esophageal cancer was explored. The effects of MTE on the proliferation of human esophageal cancer cells (KYSE150 and Eca-109) were investigated by the MTT assay, the BrdU (bromodeoxyuridine) incorporation immunofluorescence assay, and flow cytometric analysis. MTE inhibited cell proliferation through inducing G0/G1 cell cycle arrest in KYSE150 and Eca-109. Western blot analysis was employed to determine protein levels in the MTE treated cells. Compared with the control cells, the expression levels of the cell cycle regulatory proteins cyclin D1/D2/D3, cyclin E1, CDK2/4/6 (CDK: cyclin dependent kinase), and p-Rb were decreased significantly in the cells treated with MTE at 40 mg·mL(-1). In addition, MTE had an inhibitory effect on the MAPK (mitogen-activated protein kinase) signal transduction pathway, including ERK (extracellular signal-regulated kinase), JNK (c-Jun N-terminal kinase), and p38MAPK. Moreover, MTE showed little additional effects on the regulation of cyclin D1/D3, CDK4/6, and p-Rb when the ERK pathway was already inhibited by the specific ERK inhibitor U0126. In conclusion, these data suggest that MTE inhibits human esophageal cancer cell proliferation through regulation of cell cycle regulatory proteins and the MAPK signaling pathways, which is probably mediated by the inhibition of ERK activation.
Apoptosis
;
drug effects
;
Carcinoma
;
drug therapy
;
enzymology
;
physiopathology
;
Cell Line, Tumor
;
Cell Proliferation
;
drug effects
;
Drugs, Chinese Herbal
;
pharmacology
;
Esophageal Neoplasms
;
drug therapy
;
enzymology
;
physiopathology
;
Extracellular Signal-Regulated MAP Kinases
;
metabolism
;
G1 Phase Cell Cycle Checkpoints
;
drug effects
;
Humans
;
MAP Kinase Signaling System
;
drug effects
;
Marsdenia
;
chemistry
9.Apoptotic effect of cisplatin and cordycepin on OC3 human oral cancer cells.
Ying-hui CHEN ; Lyh-Jyh HAO ; Chih-peng HUNG ; Jung-wei CHEN ; Sew-fen LEU ; Bu-miin HUANG
Chinese journal of integrative medicine 2014;20(8):624-632
OBJECTIVETo evaluate apoptotic effects of cisplatin and cordycepin as single agent or in combination with cytotoxicity in oral cancer cells.
METHODSThe influences of cisplatin (2.5 μg/mL) and/or cordycepin treatment (10 or 100 μmol/L) to human OC3 oral cancer cell line were investigated by morphological observation for cell death appearance, methylthiazoletetrazolium (MTT) assay for cell viability, flow cytometry assay for cell apoptosis, and Western blotting for apoptotic protein expressions.
RESULTSData demonstrated that co-administration of cisplatin (2.5 μg/mL) and cordycepin (10 or 100 μmol/L) resulted in the enhancement of OC3 cell apoptosis compared to cisplatin or cordycepin alone treatment (24 h), respectively (P <0.05). In flow cytometry assay, percentage of cells arrested at subG1 phase with co-treatment of cordycepin and cisplatin (30%) was significantly higher than cisplatin (5%) or cordycepin (12%) alone group (P <0.05), confirming a synergistically apoptotic effect of cordycepin and cisplatin. In cellular mechanism study, co-treatment of cordycepin and cisplatin induced more stress-activated protein kinase/Jun terminal kinase (JNK), the expressions of caspase-7, and the cleavage of poly ADP-ribose polymerase (PARP) as compared to cisplatin or cordycepin alone treatment (P <0.05).
CONCLUSIONCisplatin and cordycepin possess synergistically apoptotic effect through the activation of JNK/caspase-7/PARP pathway in human OC3 oral cancer cell line.
Apoptosis ; drug effects ; Caspase 7 ; metabolism ; Cell Count ; Cell Line, Tumor ; Cell Shape ; drug effects ; Cell Survival ; drug effects ; Cisplatin ; pharmacology ; Deoxyadenosines ; pharmacology ; Drug Synergism ; G1 Phase ; drug effects ; Humans ; JNK Mitogen-Activated Protein Kinases ; metabolism ; Mouth Neoplasms ; pathology ; Phosphorylation ; drug effects ; Poly(ADP-ribose) Polymerases ; metabolism
10.Inhibitions of SphK1 inhibitor SKI II on cell cycle progression and cell invasion of hepatoma HepG2 cells.
Cai-Xia ZHANG ; Hong LIU ; Yu-Yan GONG ; Hong-Wei HE ; Rong-Guang SHAO
Acta Pharmaceutica Sinica 2014;49(2):204-208
Sphingosine kinase 1 (SphK1) plays critical roles in cell biological functions. Here we investigated the effects of SphK1 inhibitor SKI II on hepatoma HepG2 cell cycle progression and invasion. Cell survival was determined by SRB assay, cell cycle progression was assayed by flow cytometry, the ability of cell invasion was measured by Matrigel-Transwell assay and protein expression was detected by Western blotting. The results showed that SKI II markedly inhibited HepG2 cell survival in a dose-dependent manner, induced G1 phase arrest in HepG2 cell and inhibited cell invasion. SKI II markedly decreased the expressions of G1-phase-related proteins CDK2, CDK4 and Cdc2 and the levels of cell invasion-associated proteins MMP2 and MMP9. The results showed that SKI II inhibited cell cycle progression and cell invasion, implying SphK1 as a potential target for hepatoma treatment.
CDC2 Protein Kinase
;
Cell Movement
;
drug effects
;
Cell Survival
;
drug effects
;
Cyclin-Dependent Kinase 2
;
metabolism
;
Cyclin-Dependent Kinase 4
;
metabolism
;
Cyclin-Dependent Kinases
;
metabolism
;
G1 Phase
;
drug effects
;
Hep G2 Cells
;
Humans
;
Matrix Metalloproteinase 2
;
metabolism
;
Matrix Metalloproteinase 9
;
metabolism
;
Phosphotransferases (Alcohol Group Acceptor)
;
antagonists & inhibitors
;
Thiazoles
;
pharmacology

Result Analysis
Print
Save
E-mail