1.Salvianolic Acid A Protects Neonatal Cardiomyocytes Against Hypoxia/Reoxygenation-Induced Injury by Preserving Mitochondrial Function and Activating Akt/GSK-3β Signals.
Xue-Li LI ; Ji-Ping FAN ; Jian-Xun LIU ; Li-Na LIANG
Chinese journal of integrative medicine 2019;25(1):23-30
OBJECTIVE:
To investigate the effects of salvianolic acid A (SAA) on cardiomyocyte apoptosis and mitochondrial dysfunction in response to hypoxia/reoxygenation (H/R) injury and to determine whether the Akt signaling pathway might play a role.
METHODS:
An in vitro model of H/R injury was used to study outcomes on primary cultured neonatal rat cardiomyocytes. The cardiomyocytes were treated with 12.5, 25, 50 μg/mL SAA at the beginning of hypoxia and reoxygenation, respectively. Adenosine triphospate (ATP) and reactive oxygen species (ROS) levels were assayed. Cell apoptosis was evaluated by flow cytometry and the expression of cleaved-caspase 3, Bax and Bcl-2 were detected by Western blotting. The effects of SAA on mitochondrial dysfunction were examined by determining the mitochondrial membrane potential (△Ψm) and mitochondrial permeability transition pore (mPTP), followed by the phosphorylation of Akt (p-Akt) and GSK-3β (p-GSK-3β), which were measured by Western blotting.
RESULTS:
SAA significantly preserved ATP levels and reduced ROS production. Importantly, SAA markedly reduced the number of apoptotic cells and decreased cleaved-caspase 3 expression levels, while also reducing the ratio of Bax/Bcl-2. Furthermore, SAA prevented the loss of △Ψm and inhibited the activation of mPTP. Western blotting experiments further revealed that SAA significantly increased the expression of p-Akt and p-GSK-3β, and the increase in p-GSK-3β expression was attenuated after inhibition of the Akt signaling pathway with LY294002.
CONCLUSION
SAA has a protective effect on cardiomyocyte H/R injury; the underlying mechanism may be related to the preservation of mitochondrial function and the activation of the Akt/GSK-3β signaling pathway.
Adenosine Triphosphate
;
analysis
;
Animals
;
Animals, Newborn
;
Caffeic Acids
;
pharmacology
;
Cell Hypoxia
;
Cells, Cultured
;
Glycogen Synthase Kinase 3 beta
;
physiology
;
Lactates
;
pharmacology
;
Mitochondria, Heart
;
drug effects
;
physiology
;
Mitochondrial Membrane Transport Proteins
;
drug effects
;
Myocytes, Cardiac
;
drug effects
;
Proto-Oncogene Proteins c-akt
;
physiology
;
Rats
;
Rats, Sprague-Dawley
;
Reactive Oxygen Species
;
metabolism
;
Signal Transduction
;
physiology
2.Effect of telomerase activation on biological behaviors of neural stem cells in rats with hypoxic-ischemic insults.
Jun-Jie MENG ; Shi-Ping LI ; Feng-Yan ZHAO ; Yu TONG ; De-Zhi MU ; Yi QU
Chinese Journal of Contemporary Pediatrics 2017;19(2):229-236
OBJECTIVETo investigate the effect of telomerase activation on biological behaviors of neural stem cells after hypoxic-ischemic insults.
METHODSThe neural stem cells passaged in vitro were divided into four groups: control, oxygen-glucose deprivation (OGD), OGD+cycloastragenol (CAG) high concentration (final concentration of 25 μM), and OGD+CAG low concentration (final concentration of 10 μM). The latter three groups were subjected to OGD. Telomerase reverse transcriptase (TERT) expression level was evaluated by Western blot. Telomerase activity was detected by telomerase repeat amplification protocol (TRAP). Cell number and neural sphere diameter were measured under a microscope. The activity of lactate dehydrogenase (LDH) was examined by chemiluminescence. Cell proliferation rate and apoptosis were detected by flow cytometry.
RESULTSAfter OGD insults, obvious injury of neural stem cells was observed, including less cell number, smaller neural sphere, more dead cells, lower proliferation rate and decreased survival rate. In CAG-treated groups, there were higher TERT expression level and telomerase activity compared with the control group (P<0.05). In comparison with the OGD group, CAG treatment attenuated cell loss (P<0.05) and neural sphere diameter decrease (P<0.05), promoted cell proliferation (P<0.05), and increased cell survival rate (P<0.05). Low and high concentrations of CAG had similar effects on proliferation and survival of neural stem cells (P>0.05). In the normal cultural condition, CAG treatment also enhanced TERT expression (P<0.05) and increased cell numbers (P<0.05) and neural sphere diameter (P<0.05) compared with the control group.
CONCLUSIONSTelomerase activation can promote the proliferation and improve survival of neural stem cells under the state of hypoxic-ischemic insults, suggesting telomerase activators might be potential agents for the therapy of hypoxic-ischemic brain injury.
Animals ; Cell Survival ; drug effects ; Enzyme Activation ; Hypoxia-Ischemia, Brain ; etiology ; Neural Stem Cells ; drug effects ; physiology ; Rats ; Sapogenins ; pharmacology ; Telomerase ; physiology
3.Effects of Low-dose Triamcinolone Acetonide on Rat Retinal Progenitor Cells under Hypoxia Condition.
Yao XING ; Li-Jun CUI ; Qian-Yan KANG
Chinese Medical Journal 2016;129(13):1600-1606
BACKGROUNDRetinal degenerative diseases are the leading causes of blindness in developed world. Retinal progenitor cells (RPCs) play a key role in retina restoration. Triamcinolone acetonide (TA) is widely used for the treatment of retinal degenerative diseases. In this study, we investigated the role of TA on RPCs in hypoxia condition.
METHODSRPCs were primary cultured and identified by immunofluorescence staining. Cells were cultured under normoxia, hypoxia 6 h, and hypoxia 6 h with TA treatment conditions. For the TA treatment groups, after being cultured under hypoxia condition for 6 h, RPCs were treated with different concentrations of TA for 48-72 h. Cell viability was measured by cell counting kit-8 (CCK-8) assay. Cell cycle was detected by flow cytometry. Western blotting was employed to examine the expression of cyclin D1, Akt, p-Akt, nuclear factor (NF)-κB p65, and caspase-3.
RESULTSCCK-8 assays indicated that the viability of RPCs treated with 0.01 mg/ml TA in hypoxia group was improved after 48 h, comparing with control group (P < 0.05). After 72 h, the cell viability was enhanced in both 0.01 mg/ml and 0.02 mg/ml TA groups compared with control group (all P < 0.05). Flow cytometry revealed that there were more cells in S-phase in hypoxia 6 h group than in normoxia control group (P < 0.05). RPCs in S and G2/M phases decreased in groups given TA, comparing with other groups (all P < 0.05). There was no significant difference in the total Akt protein expression among different groups, whereas upregulation of p-Akt and NF-κB p65 protein expression and downregulation of caspase-3 and cyclin D1 protein expression were observed in 0.01 mg/ml TA group, comparing with hypoxia 6 h group and control group (all P < 0.05).
CONCLUSIONLow-dose TA has anti-apoptosis effect on RPCs while it has no stimulatory effect on cell proliferation.
Animals ; Apoptosis ; drug effects ; physiology ; Caspase 3 ; metabolism ; Cell Cycle ; drug effects ; physiology ; Cell Hypoxia ; drug effects ; physiology ; Cell Proliferation ; drug effects ; physiology ; Cell Survival ; drug effects ; physiology ; Cells, Cultured ; Cyclin D1 ; metabolism ; NF-kappa B ; metabolism ; Proto-Oncogene Proteins c-akt ; metabolism ; Rats ; Rats, Sprague-Dawley ; Retina ; cytology ; Stem Cells ; cytology ; drug effects ; Triamcinolone Acetonide ; pharmacology
4.Calpain mediated pulmonary vascular remodeling in hypoxia induced pulmonary hypertension.
Weifang ZHANG ; Tiantian ZHU ; Aizhen XIONG ; Xiaoyue GE ; Ruilai XU ; Shegui LU ; Changping HU
Journal of Central South University(Medical Sciences) 2016;41(9):929-936
OBJECTIVE:
To explore the role of calpain in pulmonary vascular remodeling in hypoxia-induced pulmonary hypertension and the underlying mechanisms.
METHODS:
Sprague-Dawley rats were randomly divided into the hypoxia group and the normoxia control group. Right ventricular systolic pressure (RVSP) and mean pulmonary artery pressure (mPAP) were monitored by a method with right external jugular vein cannula. Right ventricular hypertrophy index was presented as the ratio of right ventricular weight to left ventricular weight (left ventricle plus septum weight). Levels of calpain-1, -2 and -4 mRNA in pulmonary artery were determined by real-time PCR. Levels of calpain-1, -2 and -4 protein were determined by Western blot. Primary rat pulmonary arterial smooth muscle cells (PASMCs) were divided into 4 groups: a normoxia control group, a normoxia+MDL28170 group, a hypoxia group and a hypoxia+MDL28170 group. Cell proliferation was detected by MTS and flow cytometry. Levels of Ki-67 and proliferating cell nuclear antigen (PCNA) mRNA were determined by real-time PCR.
RESULTS:
RVSP, mPAP and right ventricular remodeling index were significantly elevated in the hypoxia group compared to those in the normoxia group. In the hypoxia group, pulmonary vascular remodeling was significantly developed, accompanied by up-regulation of calpain-1, -2 and -4. MDL28170 significantly inhibited hypoxia-induced proliferation of PASMCs concomitant with the suppression of Ki-67 and PCNA mRNA expression.
CONCLUSION
Calpain mediates vascular remodeling via promoting proliferation of PASMCs in hypoxia-induced pulmonary hypertension.
Animals
;
Calpain
;
genetics
;
physiology
;
Cell Proliferation
;
Dipeptides
;
physiology
;
Hypertension, Pulmonary
;
chemically induced
;
genetics
;
physiopathology
;
Hypertrophy, Right Ventricular
;
Hypoxia
;
Ki-67 Antigen
;
drug effects
;
Myocytes, Smooth Muscle
;
physiology
;
Proliferating Cell Nuclear Antigen
;
drug effects
;
Pulmonary Artery
;
Rats
;
Rats, Sprague-Dawley
;
Real-Time Polymerase Chain Reaction
;
Up-Regulation
;
Vascular Remodeling
;
genetics
;
physiology
5.Resveratrol Inhibits Hypoxia-Induced Vascular Endothelial Growth Factor Expression and Pathological Neovascularization.
Christopher Seungkyu LEE ; Eun Young CHOI ; Sung Chul LEE ; Hyoung Jun KOH ; Joon Haeng LEE ; Ji Hyung CHUNG
Yonsei Medical Journal 2015;56(6):1678-1685
PURPOSE: To investigate the effects of resveratrol on the expression of hypoxia-inducible factor 1alpha (HIF-1alpha) and vascular endothelial growth factor (VEGF) in human adult retinal pigment epithelial (ARPE-19) cells, and on experimental choroidal neovascularization (CNV) in mice. MATERIALS AND METHODS: ARPE-19 cells were treated with different concentrations of resveratrol and then incubated under hypoxic conditions with subsequent evaluation of cell viability, expression of HIF-1alpha, and expression of VEGF. The effects of resveratrol on the synthesis and degradation of hypoxia-induced HIF-1alpha were evaluated using inhibitors of the PI3K/Akt/mTOR and the ubiquitin proteasome pathways. In animal studies, CNV lesions were induced in C57BL/6 mice by laser photocoagulation. After 7 days of oral administration of resveratrol or vehicle, which began one day after CNV induction, image analysis was used to measure CNV areas on choroidal flat mounts stained with isolectin IB4. RESULTS: In ARPE-19 cells, resveratrol significantly inhibited HIF-1alpha and VEGF in a dose-dependent manner, by blocking the PI3K/Akt/mTOR signaling pathway and by promoting proteasomal HIF-1alpha degradation. In mice experiments, orally administered resveratrol significantly inhibited CNV growth in a dose-dependent manner. CONCLUSION: Resveratrol may have therapeutic value in the management of diseases involving pathological neovascularization.
Adult
;
Animals
;
Anoxia/metabolism/physiopathology
;
Cell Survival/drug effects
;
Choroidal Neovascularization/*metabolism/pathology
;
Humans
;
Hypoxia-Inducible Factor 1, alpha Subunit/*drug effects/metabolism
;
Mice
;
Mice, Inbred C57BL
;
Phosphatidylinositol 3-Kinases/antagonists & inhibitors/*physiology
;
Proteasome Endopeptidase Complex
;
Proto-Oncogene Proteins c-akt/antagonists & inhibitors/*physiology
;
Retinal Pigment Epithelium/*drug effects/metabolism
;
Signal Transduction
;
Stilbenes/administration & dosage/*pharmacology
;
TOR Serine-Threonine Kinases/antagonists & inhibitors/*physiology
;
Ubiquitin
;
Vascular Endothelial Growth Factor A/*drug effects/metabolism
6.Regulatory effects of AT₁R-TRAF6-MAPKs signaling on proliferation of intermittent hypoxia-induced human umbilical vein endothelial cells.
Jin SHANG ; Xue-Ling GUO ; Yan DENG ; Xiao YUAN ; Hui-Guo LIU
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(4):495-501
Endothelial dysfunction induced by intermittent hypoxia (IH) participates in obstructive sleep apnea syndrome (OSAS)-associated cardiovascular disorders. Myeloid differentiation primary response 88 (MyD88) and tumor necrosis factor receptor-associated factor 6 (TRAF6) regulate numerous downstream adaptors like mitogen-activated protein kinases (MAPKs) and the subsequent oxidative stress and inflammatory responses. This study aimed to characterize the role of MyD88/TRAF6 in IH-treated cell function and its associated signaling. Human umbilical vein endothelial cells (HUVECs) were randomly exposed to IH or normoxia for 0, 2, 4 and 6 h. Western blotting was used to detect the expression pattern of target gene proteins [angiotensin 1 receptor (AT1R), p-ERK1/2, p-p38MAPK, MyD88 and TRAF6], and the relationships among these target genes down-regulated by the corresponding inhibitors were studied. Finally, the influence of these target genes on proliferation of HUVECs was also assessed by EdU analysis. Protein levels of AT1R, TRAF6 and p-ERK1/2 were increased after IH exposure, with a slight rise in MyD88 and a dynamic change in p-p38MAPK. The down-regulation of TRAF6 by siRNA reduced ERK1/2 phosphorylation during IH without any effects on AT1R. Blockade of AT1R with valsartan decreased TRAF6 and p-ERK1/2 protein expression after IH exposure. ERK1/2 inhibition with PD98059 suppressed only AT1R expression. IH promoted HUVECs proliferation, which was significantly suppressed by the inhibition of TRAF6, AT1R and ERK1/2. The findings demonstrate that TRAF6 regulates the proliferation of HUVECs exposed to short-term IH by modulating cell signaling involving ERK1/2 downstream of AT1R. Targeting the AT1R-TRAF6-p-ERK1/2 signaling pathway might be helpful in restoring endothelial function.
Cell Hypoxia
;
Cell Proliferation
;
Cells, Cultured
;
Gene Expression Regulation
;
Human Umbilical Vein Endothelial Cells
;
physiology
;
Humans
;
MAP Kinase Signaling System
;
drug effects
;
Phosphorylation
;
Receptor, Angiotensin, Type 1
;
genetics
;
metabolism
;
TNF Receptor-Associated Factor 6
;
genetics
;
metabolism
;
Valsartan
;
pharmacology
7.Recent highlights of experimental research for inhibiting tumor growth by using Chinese medicine.
Xi-ran HE ; Shu-yan HAN ; Ping-ping LI
Chinese journal of integrative medicine 2015;21(10):727-732
To give an overview of contemporary experimental research using Chinese medicine (CM) for the treatment of cancer. As an integral part of mainstream medicine in the People's Republic of China, CM emphasizes improvements in holistic physical condition instead of merely killing tumor cells, which is consistent with the current medical model that advocates patient-oriented treatment. Great progress has been made in experimental research, and the principle aspects include anti-tumor angiogenesis, inducing apoptosis and differentiation, reversing multidrug resistance, and improving immune function. As a current hot topic in cancer research, tumor microenvironment (TME) highlights the mutual and interdependent interaction between tumor cells and their surrounding tissues, and the CM treatment concept bears a striking resemblance to it. To date, primary points of TME include extracellular matrix remodeling, inflammation, hypoxia, and angiogenesis, but trials using CM with a focus on TME are rare. Despite considerable recent development, experimental research on CM for solving cancer issues appears insufficient. Greater efforts in this field are urgently needed.
Apoptosis
;
drug effects
;
Autophagy
;
drug effects
;
Cell Differentiation
;
drug effects
;
Cell Hypoxia
;
drug effects
;
Drug Resistance, Multiple
;
drug effects
;
Immunomodulation
;
Inflammation Mediators
;
pharmacology
;
Medicine, Chinese Traditional
;
Neoplasms, Experimental
;
Research
;
Tumor Microenvironment
;
drug effects
;
physiology
8.Effect of puerarin on hypoxia induced proliferation of PASMCs by regulating reactive oxygen.
Xiao-dan ZHANG ; Li-wei WANG ; Shu-jing WANG ; Da-ling ZHU ; Yan-nan YANG ; Jie-jing SHENG ; Sha-sha SONG
China Journal of Chinese Materia Medica 2015;40(15):3027-3033
To discuss the effect of puerarin (Pue) on the proliferation of hypoxia-induced pulmonary artery smooth muscle cells (PASMCs) and discuss whether its mechanism is achieved by regulating reactive oxygen. PASMCs of primarily cultured rats (2-5 generations) were selected in the experiment. MTT, Western blot, FCM and DCFH-DA were used to observe Pue's effect the proliferation of PASMCs. The Western blot was adopted to detect whether ROS participated in Pue's effect in inhibiting PASMC proliferation. The PASMCs were divided into five groups: the normoxia group, the hypoxia group, the hypoxia + Pue group, the hypoxia + Pue + Rotenone group and the hypoxia + Rotenone group, with Rotenone as the ROS blocker. According to the results, under the conditions of normoxia, Pue had no effect on the PASMC proliferation; But, under the conditions of hypoxia, it could inhibit the PASMC proliferation; Under the conditions of normoxia and hypoxia, Pue had no effect on the expression of the tumor necrosis factor-α (TNF-α) among PASMCs, could down-regulate the expression of hypoxia-induced cell cycle protein Cyclin A and proliferative nuclear antigen (PCNA). DCFH-DA proved Pue could reverse ROS rise caused by hypoxia. Both Rotenone and Pue could inhibit the up-regulated expressions of HIF-1α, Cyclin A, PCNA caused by anoxia, with a synergistic effect. The results suggested that Pue could inhibit the hypoxia-induced PASMC proliferation. Its mechanism may be achieved by regulating ROS.
Animals
;
Cell Cycle
;
drug effects
;
Cell Proliferation
;
drug effects
;
Cells, Cultured
;
Hypoxia
;
pathology
;
Isoflavones
;
pharmacology
;
Male
;
Myocytes, Smooth Muscle
;
drug effects
;
physiology
;
Proliferating Cell Nuclear Antigen
;
analysis
;
Pulmonary Artery
;
cytology
;
drug effects
;
Rats
;
Rats, Wistar
;
Reactive Oxygen Species
;
metabolism
9.Scutellarin attenuates endothelium-dependent aasodilation impairment induced by hypoxia reoxygenation, through regulating the PKG signaling pathway in rat coronary artery.
Ya-Juan CHEN ; Lei WANG ; Guang-Yu ZHOU ; Xian-Lun YU ; Yong-Hui ZHANG ; Na HU ; Qing-Qing LI ; Chen CHEN ; Chen QING ; Ying-Ting LIU ; Wei-Min YANG
Chinese Journal of Natural Medicines (English Ed.) 2015;13(4):264-273
Scutellarin (SCU), a flavonoid from a traditional Chinese medicinal plant. Our previous study has demonstrated that SCU relaxes mouse aortic arteries mainly in an endothelium-depend-ent manner. In the present study, we investigated the vasoprotective effects of SCU against HR-induced endothelial dysfunction (ED) in isolated rat CA and the possible mechanisms involving cyclic guanosine monophosphate (cGMP) dependent protein kinase (PKG). The isolated endothelium-intact and endothelium-denuded rat CA rings were treated with HR injury. Evaluation of endothelium-dependent and -independent vasodilation relaxation of the CA rings were performed using wire myography and the protein expressions were assayed by Western blotting. SCU (10-1 000 μmol·L(-1)) could relax the endothelium-intact CA rings but not endothelium-denuded ones. In the intact CA rings, the PKG inhibitor, Rp-8-Br-cGMPS (PKGI-rp, 4 μmol·L(-1)), significantly blocked SCU (10-1 000 μmol·L(-1))-induced relaxation. The NO synthase (NOS) inhibitor, NO-nitro-L-arginine methylester (L-NAME, 100 μmol·L(-1)), did not significantly change the effects of SCU (10-1 000 μmol·L(-1)). HR treatment significantly impaired ACh-induced relaxation, which was reversed by pre-incubation with SCU (500 μmol·L(-1)), while HR treatment did not altered NTG-induced vasodilation. PKGI-rp (4 μmol·L(-1)) blocked the protective effects of SCU in HR-treated CA rings. Additionally, HR treatment reduced phosphorylated vasodilator-stimulated phosphoprotein (p-VASP, phosphorylated product of PKG), which was reversed by SCU pre-incubation, suggesting that SCU activated PKG phosphorylation against HR injury. SCU induces CA vasodilation in an endothelium-dependent manner to and repairs HR-induced impairment via activation of PKG signaling pathway.
Animals
;
Apigenin
;
pharmacology
;
Cell Adhesion Molecules
;
drug effects
;
Cell Hypoxia
;
Coronary Vessels
;
drug effects
;
Cyclic GMP
;
analogs & derivatives
;
metabolism
;
pharmacology
;
Cyclic GMP-Dependent Protein Kinases
;
Glucuronates
;
pharmacology
;
Microfilament Proteins
;
drug effects
;
NG-Nitroarginine Methyl Ester
;
metabolism
;
pharmacology
;
Phosphoproteins
;
drug effects
;
Rats
;
Rats, Sprague-Dawley
;
Reperfusion Injury
;
complications
;
physiopathology
;
Signal Transduction
;
drug effects
;
Thionucleotides
;
metabolism
;
pharmacology
;
Vasodilation
;
drug effects
;
physiology
10.The role of adiponectin in the production of IL-6, IL-8, VEGF and MMPs in human endothelial cells and osteoblasts: implications for arthritic joints.
Yeon Ah LEE ; Hye In JI ; Sang Hoon LEE ; Seung Jae HONG ; Hyung In YANG ; Myung CHUL YOO ; Kyoung Soo KIM
Experimental & Molecular Medicine 2014;46(1):e72-
This study was performed to evaluate the contribution of adiponectin to the production of interleukin (IL)-6, IL-8, vascular endothelial growth factor (VEGF), matrix metalloproteinase (MMP)-1 and MMP-13 in human endothelial cells and osteoblasts in arthritic joints. Cultured human umbilical vascular endothelial cells (HUVECs) and osteoblasts were stimulated with adiponectin (1 or 10 mug ml-1) or IL-1beta (0.1 ng ml-1) in the presence or absence of hypoxia for 24 h. The protein expression patterns were examined by analyzing culture supernatants using the enzyme-linked immunosorbent assay (ELISA). Adiponectin significantly stimulated the production of VEGF, MMP-1 and MMP-13 in osteoblasts but not in endothelial cells, whereas it significantly stimulated the production of IL-6 and IL-8 in both endothelial cells and osteoblasts. The increase in VEGF production induced by adiponectin was significantly greater than that induced by IL-1beta. The production of IL-6 and IL-8 in adiponectin-stimulated endothelial cells was approximately 10-fold higher than that in IL-1beta-stimulated endothelial cells; in osteoblasts, adiponectin-induced IL-6 and IL-8 secretion was approximately twofold higher than that induced by IL-1beta. In addition, IL-8 production in endothelial cells was approximately sevenfold higher than in osteoblasts. However, IL-6 levels were similar between the two cell types, suggesting that adiponectin may be involved in the production of IL-8 in endothelial cells, which may have an important role in neutrophil recruitment to arthritic joints. Furthermore, the increases in protein expression induced by adiponectin were differentially regulated by hypoxia. In conclusion, adiponectin has a more important role than does IL-1beta in the production of mediators that drive synovitis and joint destruction in endothelial cells and osteoblasts at physiological concentrations.
Adiponectin/pharmacology/*physiology
;
Arthritis, Rheumatoid/metabolism
;
Cell Hypoxia
;
Cell Line
;
Human Umbilical Vein Endothelial Cells/drug effects/*metabolism
;
Humans
;
Interleukin-6/genetics/*metabolism
;
Interleukin-8/genetics/*metabolism
;
Matrix Metalloproteinase 1/genetics/*metabolism
;
Osteoblasts/drug effects/*metabolism
;
Vascular Endothelial Growth Factor A/genetics/*metabolism

Result Analysis
Print
Save
E-mail