Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add filters








Year range
1.
Journal of Xi'an Jiaotong University(Medical Sciences) ; (6): 227-231, 2018.
Article in Chinese | WPRIM | ID: wpr-698232

ABSTRACT

Objective To investigate the possible molecular mechanism for alpha(α)-mangostin's inhibition of the proliferation and apoptosis of human gastric cancer cells.Methods Human gastric adenocarcinoma SGC7901 cell line was treated with α-mangostin.CCK8 method was used to measure the viability of SGC7901 cells.The effect of α-mangostin on apoptosis and cell cycle was determined by immune fluorescence and flow cytometry.The expression of the relevant proteins was detected using Western blot.The shapiro-wilk test was performed for evaluation of deviation from normality.Normally distributed data was analyzed with one-way ANOVA.Welch test was used in data with heterogeneity of variance and multiple compared by Games-Howell test after that.Results CCK8 results showed that cell viability differed significantly among groups treated with different concentrations of α-mangostin(10,15,20,25,and 30 μmol/L)(P<0.05).QPCR data showed that the concentration of α-mangostin was positively correlated with mRNA level of LC 3 but not caspase protein(r=0.976,P<0.05).In 15 μmol/L but not 10 μmol/L α-mangostin treatment system,the autophagy inhibitors 3-MA(10 μmol/L),bafilomycin A(10 μmol/L)and LY294002(10 μmol/L)could significantly alleviate α-mangostin's killing effect on SGC7901 cells(P<0.05).Conclusion The anti-tumor effects of α-mangostin against human gastric adenocarcinoma cells in vitro can be partly attributed to apoptosis,autophagy and arresting cell phase.

2.
Journal of Southern Medical University ; (12): 866-868, 2017.
Article in Chinese | WPRIM | ID: wpr-360172

ABSTRACT

Small intestinal hemangioma is a rare condition that can be divided histologically into capillary, cavernous or mixed types, among which the cavernous type is the most common. Here we report a case of small intestinal cavernous hemangioma with chronic hemorrhage in 44-year-old man. The patient complained of weakness and dizziness for 2 years that aggravated 1 month before admission accompanied by intermittent melena. Laboratory tests suggest severe anemia, and computed tomography, gastroscopy and colonoscopy all revealed signs of anemia. Capsule endoscopy detected small intestinal erosions, bleeding lesions and prominent neoplasms. An exploratory laparotomy was performed, in which the segment of the jejunum with lesions was resected. Pathological examination of the resected jejunum identified the neoplasm as cavernous hemangioma of the small intestine, which was the cause of severe anemia.

3.
Journal of Experimental Hematology ; (6): 154-158, 2012.
Article in Chinese | WPRIM | ID: wpr-331000

ABSTRACT

The aim of this study was to explore the effect of mesenchymal stem cell (MSC) conditioned medium (MSC-CM) on proliferation, migration and adhesion of human umbilical vein endothelial cell (CRL1730) and its mechanism. Isolation and purification of MSC were performed with the classic adhering method, the surface markers (CD29, CD90, CD45 and CD34) in MSC were detected by flow cytometry. MSC were treated and cultured for 3 d, the MSC-CM or MSC overexpressing stem cell-derived factor-1 (SDF-1) conditioned medium (Ad-SDF-1-MSC-CM) were collected. Subsequently, CRL1730 cells were treated respectively with 2% FBS-DMEM, 15% FBS-DMEM (control group), MSC-CM or Ad-SDF-1-MSC-CM for 24 h, the proliferation of CRL1730 cells was detected by MTT method. CRL1730 cell migration in vitro was performed by using wound healing system. The adhesion ability of CRL1730 cells was analyzed by microscope. The results indicated that the CRL1730 cells treated with Ad-SDF-1-MSC-CM showed greater proliferative capacity than CRL1730 cells treated with MSC-CM. While adding with AMD3100 5 µmol/L, the blocker of CXCR4, the CRL1730 proliferation mediated by Ad-SDF-1-MSC-CM was significantly reduced. Meanwhile, compared with MSC-CM, Ad-SDF-1-MSC-CM had greater effects for promoting CRL1730 migration and enhancing adhesion ability of CRL1730 cells, these effects were significantly inhibited by AMD3100. It is concluded that MSC-CM promotes the migration and adhesion ability of CRL1730 cells through SDF-1 expressed by MSC.


Subject(s)
Humans , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Culture Media, Conditioned , Human Umbilical Vein Endothelial Cells , Cell Biology , Mesenchymal Stem Cells , Cell Biology
4.
Journal of Southern Medical University ; (12): 1697-1700, 2011.
Article in Chinese | WPRIM | ID: wpr-333834

ABSTRACT

<p><b>OBJECTIVE</b>To observe the effect of vascular endothelial growth factor (VEGF) on bone marrow-derived mesenchymal stem cell (MSC) proliferation and explore the signaling mechanism involved.</p><p><b>METHODS</b>MSC culture was performed following the classical whole bone marrow adhering method. The characteristics of MSC were identified by induction of multi-lineage differentiation and flow cytometry for surface marker analysis (CD34, CD45, CD29, and CD90). Following the addition of 50 nmol/L wortmannin, 50 µmol/L PD98059, 30 µmol/L SB203580, 10 µmol/L H89, 20 µmol/L Y27632, 1 µmol/L rapamycin, 10 µmol/L straurosporine, 6 nmol/L Go6976, or 50 µmol/L Pseudo Z inhibitors in the cell culture, the MSC were treated with 20 ng/ml VEGF and the changes of the cell proliferation rate was measured with MTT assay.</p><p><b>RESULTS</b>Cultured MSC were capable of multi-linage differentiation and did not express VEGF-R, CD29 or CD90. Treatment with 20 ng/ml VEGF obviously promoted MSC proliferation, and this effect was inhibited partially by p38 mitogen-activated protein kinase (MAPK) inhibitor rapamycin, PD98059, SB203580, Go6976, and straurosporine.</p><p><b>CONCLUSIONS</b>VEGF promotes MSC proliferation in close relation to the AKT-PKC pathway, in which PKC signal pathway may play the central role.</p>


Subject(s)
Animals , Female , Male , Rats , Bone Marrow Cells , Cell Biology , Cell Proliferation , Cells, Cultured , Mesenchymal Stem Cells , Cell Biology , Protein Kinase C , Metabolism , Rats, Sprague-Dawley , Signal Transduction , Vascular Endothelial Growth Factor A , Pharmacology
5.
Journal of Experimental Hematology ; (6): 1292-1296, 2010.
Article in Chinese | WPRIM | ID: wpr-332374

ABSTRACT

In order to explore the effect of VEGF on mesenchymal stem cell (MSC) proliferation and its possible signal transduction mechanism, MSC culture was performed with the classical bone marrow adhering method; characteristics of passage 3 rat MSC (P3MSC) was identified through multi-differentiation and surface marker assay (CD34, CD45, CD90, CD29); P3MSC were treated with 20 ng/ml VEGF, and the effect of VEGF on the MSC proliferation was measured during 12, 36 and 72 hours by MTT assay. Subsequently, P3MSC were treated with extracellular-signal regulated kinase (ERK1/2) inhibitor PD98059 (50 µmol/L) or p38 mitogen-activated protein kinase (p38MAPK) inhibitor SB203580 (30 µmol/L) for 30 minutes, the culture medium was replaced with new medium including 20 ng/ml VEGF. After 72 hours, the effect of PD98059 or SB203580 on MSC proliferation mediated by VEGF was measured by MTT assay. The result showed that the cultured MSC expressed PDGFR-α, PDGFR-β and NRP1, but did not express VEGF-R (Flk1 and Flt1). The MSC had the multi-differentiation ability and displayed the characteristics of CD90+ (96.7%), CD29+ (94.6%), CD34- (0.79%) and CD45- (0.84%). The MSC proliferation rate increased gradually with prolonging of the functioning time of 20 ng/ml VEGF, and MSC proliferation rate may reach to maximum value after treating with 20 ng/ml VEGF for 72 hours. The effect of VEGF on MSC proliferation was found to be abolished, even was under level of control group after treating with PD98059 or SB203580 for 30 minutes. Furthermore, the inhibitory effect of PD98059 on MSC proliferation was obviously higher than that of SB203580. It is concluded that the VEGF can promote MSC proliferation, and its possible mechanism may relate to ERK1/2 pathway.


Subject(s)
Animals , Rats , Bone Marrow Cells , Cell Biology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases , Metabolism , Flavonoids , Pharmacology , Imidazoles , Pharmacology , Mesenchymal Stem Cells , Cell Biology , Pyridines , Pharmacology , Rats, Sprague-Dawley , Signal Transduction , Vascular Endothelial Growth Factors , Pharmacology
6.
Journal of Southern Medical University ; (12): 38-42, 2010.
Article in Chinese | WPRIM | ID: wpr-269632

ABSTRACT

<p><b>OBJECTIVE</b>To explore the effect of adenovirus-mediated human stromal cell-derived factor-1alpha (hSDF-1alpha) on ventricular remodeling in rats with myocardial infarction.</p><p><b>METHODS</b>A recombinant adenoviral plasmid containing hSDF-1alpha cDNA was constructed using homologous recombination in bacteria and the recombinant adenovirus particles expressing hSDF-1alpha (AdV-SDF-1) were prepared. In rat models of myocardial infarction induced by left anterior descending artery occlusion, 1x10(10) PFU AdV-SDF-1 or PFU AdV-LacZ were injected at multiple sites into the infarcted myocardium 1 h after the operation, using 200 l cell-free PBS as the control. Four weeks after the injection, the cardiac function of the rats was analyzed, and the heart tissues were taken after the measurement of hemodynamics. On serial frozen sections, histological observation and morphometric measurement were carried out using a microscopic image analysis system, and the expression of hSDF-1alpha was detected by immunocytochemistry.</p><p><b>RESULTS</b>Four weeks after AdV-SDF-1 injection, the myocardium in the infracted area showed significantly higher expression rates of hSDF-1alpha. The injection resulted in a obvious reduction in the infarct size and collagen content and a marked increase in the left ventricle wall, and the rats showed improved cardiac functions.</p><p><b>CONCLUSION</b>SDF-1alpha can improve the cardiac structure and function in rats with myocardial infarction by inhibiting collagen synthesis and deposition in the infarcted area.</p>


Subject(s)
Animals , Female , Male , Rats , Adenoviridae , Genetics , Metabolism , Chemokine CXCL12 , Genetics , Gene Transfer Techniques , Genetic Vectors , Genetics , Myocardial Infarction , Therapeutics , Rats, Sprague-Dawley , Recombinant Proteins , Genetics , Transfection , Ventricular Remodeling
7.
Journal of Experimental Hematology ; (6): 404-407, 2009.
Article in Chinese | WPRIM | ID: wpr-302123

ABSTRACT

The aim of this study was to explore the difference of MSC migration mediated by SDF-1/CXCR4 axis through Boyden chamber in vitro migration assay. The SDF-1 density-dependence of MSC migration was observed. Subsequently, the effects of different blocking agents on hSDF-MSC migration were observed after MSC were treated with 50 nmol/L wortmannin, 10 micromol/L LY294002, 50 micromol/L PD98059, 10 micromol/L U73122, 126 micromol/L AMD3100 and 50 nmol/L verapamil respectively. The results showed the efficiency of MSC migration increased gradually with the increasing of hSDF-1 density. And after MSCs treatment with 50 nmol/L wortmannin, 10 micromol/L LY294002, 50 micromol/L PD98059, 10 micromol/L U73122 and 126 micromol/L AMD3100 respectively, the ability of MSC migration decreased. The ability of MSCs migration obviously decreased when MSCs were treated with U73122, AMD3100. It is concluded that the SDF-1/CXCR4-mediated MSC migration may be related to mitogen-activated protein kinase (MAPK), phosphatidylinositol phospholipase C (PI-PLC) and protein kinase (PKC) signal pathways.


Subject(s)
Animals , Rats , Bone Marrow Cells , Cell Biology , Cell Movement , Flavonoids , Pharmacology , Mesenchymal Stem Cells , Cell Biology , Protein Kinase C , Metabolism , Rats, Wistar , Receptors, CXCR4 , Metabolism , Signal Transduction
8.
Journal of Southern Medical University ; (12): 2429-2432, 2009.
Article in Chinese | WPRIM | ID: wpr-325098

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the transduction efficiency of purified PEP-1-CAT fusion protein into rat heart and the protective effect of the fusion protein against myocardial ischemia-reperfusion injury.</p><p><b>METHODS</b>PEP-1-CAT or CAT (500 microg) was injected in SD rats via the caudal vein, using normal saline as the control, and the hearts were harvested at 0.5, 1, 2, 4, 8, and 24 h after the injection. The transduction efficiency was evaluated by immunofluorescence technique, and the CAT activity was measured. Forty rats were randomized into 5 groups, namely the sham-operated group, ischemia-reperfusion group, and 3 PEP-1-CAT -treated groups (100, 300, and 500 microg). The left main coronary artery was occluded for 1 h followed by a 2-h reperfusion, and at the end of reperfusion, serum LDH and CK and MDA content in the myocardium were measured.</p><p><b>RESULTS</b>No green fluorescence was observed in saline group or CAT group. Bright green fluorescence was observed in PEP-1-CAT groups at different time points, most conspicuous at 8 h. No significant difference in CAT activity was found between CAT group and saline group (P>0.05); with the lapse of time, CAT activity in PEP-1-CAT group increased gradually, reaching the peak level at 8 h, which was 4.2 folds of that in the saline group. LDH ,CK and MDA were significantly lower in PEP-1-CAT- groups than in ischemia-reperfusion group (P<0.01).</p><p><b>CONCLUSION</b>PEP-1 can mediate the transduction of CAT in rat heart in a time-dependent manner, and PEP-1-CAT preconditioning provides a protective effect against ischemia- reperfusion injury in rats.</p>


Subject(s)
Animals , Male , Rats , Catalase , Metabolism , Pharmacology , Cysteamine , Metabolism , Pharmacology , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury , Metabolism , Pathology , Peptides , Metabolism , Pharmacology , Random Allocation , Rats, Sprague-Dawley , Recombinant Fusion Proteins , Pharmacology , Transduction, Genetic
9.
Chinese Journal of Cardiology ; (12): 268-274, 2009.
Article in Chinese | WPRIM | ID: wpr-294736

ABSTRACT

<p><b>OBJECTIVE</b>The transduction efficiency of the purified PEP-1-SOD1 fusion protein and the effects of PEP-1-SOD1 fusion protein on ischemia reperfusion injury in the isolated perfused rat hearts were investigated.</p><p><b>METHODS</b>The constructed pET15b-SOD1 and pET15b-PEP-1-SOD1 were transformed into BL21 (DE3) for expression and purification of SOD1 and PEP-1-SOD1, respectively. Isolated perfused rat hearts were subjected to 60 min of global ischemia and 30 min of reperfusion and treated with vehicle, 100 micromol/L SOD1 and 25, 50, 100 micromol/L PEP-1-SOD1, respectively. The transduction efficiency was evaluated with immunofluorescent microscopy and Western blot. The enzyme activity of the transduced PEP-1-SOD1 was measured with commercial SOD detection kit. The MDA content in myocardial tissue and the CK activity in coronary exudate at 15 min after reperfusion were also measured. Cardiomyocyte apoptosis was detected with TUNEL. The infarct size was determined in isolated hearts 60 min after reperfusion with TTC staining.</p><p><b>RESULTS</b>Immunofluorescent microscopy and Western blot demonstrated PEP-1-SOD1 was transduced into myocardial tissue in a dose-dependent manner, whereas SOD1 could not be detected in SOD1 group. SOD activity in control, SOD1 group, 25, 50, 100 micromol/L PEP-1-SOD1 groups was (10.06 +/- 0.77) U/mg prot, (10.59 +/- 0.71) U/mg prot, (32.29 +/- 1.42) U/mg prot, (43.16 +/- 1.16) U/mg prot, (55.14 +/- 1.59) U/mg prot, respectively. MDA content in corresponding groups was (1.48 +/- 0.19) nmol/mg prot, (1.39 +/- 0.11) nmol/mg prot, (1.01 +/- 0.14) nmol/mg prot, (0.73 +/- 0.13) nmol/mg prot, (0.50 +/- 0.06) nmol/mg prot, respectively. CK activity in corresponding groups was (1.73 +/- 0.58) U/mg prot,(1.68 +/- 0.14) U/mg prot,(1.40 +/- 0.28) U/mg prot,(0.97 +/- 0.39) U/mg prot, (0.61 +/- 0.56) U/mg prot, respectively. Cardiomyocyte apoptotic index in corresponding groups was (17.25 +/- 0.75)%, (16.63 +/- 1.07)%, (11.50 +/- 0.57) U/mg prot, (6.50 +/- 0.63) U/mg prot, (4.13 +/- 0.52)%, repectively. The percentage of myocardial infarction area was (55.13 +/- 2.18)%, (52.13 +/- 2.59)%, (33.88 +/- 2.06)%, (25.50 +/- 2.16)%, (15.38 +/- 1.14)%, respectively. Compared with control group and SOD1 group, all P < 0.01 These results demonstrated the enzyme activity of the transduced PEP-1-SOD1 was significantly increased in a dose-dependent manner and the MDA content, CK activity, the cardiomyocyte apoptotic index and the infarct size was decreased siginificantly in PEP-1-SOD1 pretreatment groups compared with SOD1 group.</p><p><b>CONCLUSION</b>The native, biologically active form of PEP-SOD1 fusion protein could be effectively transduced into the isolated rat hearts subjecting ischemia reperfusion injury in a dose-dependent manner. The transduced PEP-1-SOD1 has protective effects on ischemia reperfusion injury in the isolated rat hearts.</p>


Subject(s)
Animals , Rats , Apoptosis , Heart , Myocardial Infarction , Myocardial Reperfusion Injury , Metabolism , Myocardium , Metabolism , Rats, Sprague-Dawley , Reperfusion Injury
10.
West China Journal of Stomatology ; (6): 92-99, 2009.
Article in Chinese | WPRIM | ID: wpr-248301

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the intensity changes of different thickness ratios of Empress II glass ceramic and GI- II glass-infiltrated alumina ceramic before and after Hertzian contact cyclic fatigue.</p><p><b>METHODS</b>Disk-shaped specimens of different thickness ratios of Empress II glass ceramic and GI-II glass-infiltrated alumina ceramic were respectively fabricated. Hertzian contact technique was imposed on the specimens. Critical loads of the specimens before and after 10(5) cycles loading were recorded.</p><p><b>RESULTS</b>The average values of critical loads of all specimens reduced significantly after cycles loading (P < 0.05), and critical loads declined with diminishing thickness of the core ceramic. The critical load of GI-II glass-infiltrated alumina ceramic was significantly higher than Empress II glass ceramic before and after cycle loading(P < 0.05).</p><p><b>CONCLUSION</b>Cycle loading can reduce the strengths of the two kinds of dental ceramic, and the latter is better than the former in the resistance to fracture and cyclic fatigue. Critical loads of the two kinds of dental ceramic are mainly influenced by the core ceramic's strength and thickness.</p>


Subject(s)
Humans , Aluminum Oxide , Ceramics , Dental Porcelain , Glass , Materials Testing
11.
Acta Physiologica Sinica ; (6): 169-174, 2009.
Article in Chinese | WPRIM | ID: wpr-302465

ABSTRACT

The present study was aimed to investigate the mechanism of the granulocyte colony-stimulating factor (G-CSF) on the viability of the bone marrow mesenchymal stem cells (MSCs). MSCs were cultured by classical whole bone marrow adhering method, and the MSCs were analyzed for the cell surface differentiation markers CD34, CD133, CD90 and CD105 by flow cytometry (FCM). The ability of the MSCs to differentiate into osteocytes and adipocytes was tested in osteogenic and adipogenic mediums, separately. The effect of G-CSF (20 mug/mL) on the passage 3 MSCs viability was evaluated by MTT method, and the molecular mechanism of the G-CSF mediated effects was assayed through the pretreatment of the signal pathway inhibitors including 50 nmol/L wortmannin (phosphatidylinoesitol 3 kinase inhibitor), 50 mumol/L PD98059 [extracellular signal-regulated-kinase1/2 (ERK1/2) inhibitor], 30 mumol/L SB203580 (p38 mitogen-activated protein kinase inhibitor), 10 mumol/L H89 (protein kinase A inhibitor), 20 mumol/L Y27632 (Rho kinase inhibitor), 1 mumol/L rapamycin [mammalian target of rapamycin (mTOR) inhibitor], 10 mmol/L straurosporine [protein kinase C (PKC) inhibitor], 6 nmol/L G0697 (PKCalpha inhibitor) and 50 mumol/L Pseudo Z (PKCzeta inhibitor). Cultured passage 3 MSCs expressed CD90 and CD105 strongly, and showed the ability of multi-differentiation into osteocytes and adipocytes. G-CSF promoted the viability of MSCs, and the promotion was completely inhibited by PKC inhibitor straurosporine and partially inhibited by wortmannin, rapamycin, PD98059, SB203580 or G0697. However, its effect was not inhibited by H89, Y27632 and Pseudo Z. It is thus suggested that the promoting effect of G-CSF on MSCs viability was closely related to AKT-mTOR-PKC signal pathway, and PKC maybe the central role in the signal pathway.


Subject(s)
Animals , Humans , Bone Marrow Cells , Cell Biology , Cell Differentiation , Cell Survival , Cells, Cultured , Enzyme Inhibitors , Pharmacology , Granulocyte Colony-Stimulating Factor , Pharmacology , Hematopoietic Stem Cells , Mesenchymal Stem Cells , Cell Biology , Signal Transduction
12.
Journal of Southern Medical University ; (12): 1190-1194, 2008.
Article in Chinese | WPRIM | ID: wpr-270178

ABSTRACT

<p><b>OBJECTIVE</b>To explore the role of stromal-derived factor-1 (SDF-1) in the migration of mesenchymal stem cells (MSCs) and the underlying signal transduction mechanism.</p><p><b>METHODS</b>Rat bone marrow-derived MSCs were infected with 100 ml recombinant adenovirus containing human SDF-1alpha gene (Ad-hSDF-1alpha), and the cell migration changes were observed at 1, 2, and 3 days after the infection. Twelve hours after Ad-hSDF-1alpha transfection, the MSCs in separate cultures were treated with wortmannin (50 nmol/L), LY294002 (10 mmol/L), PD98059 (50 mmol/L), U73122 (10 mmol/L), AMD3100 (0.1 g/L), or verapamil (50 nmol/L), respectively, and the signal transduction pathways involved in MSC migration were analyzed.</p><p><b>RESULTS</b>The MSCs grew in colonies after transfection with Ad-hSDF-1alpha, but this growth pattern was substantially attenuated after treatment with wortmannin, LY294002, PD98059, U73122, AMD3100 and verapamil, among which U73122 and AMD3100 treatments resulted in the most conspicuous inhibitory effects.</p><p><b>CONCLUSION</b>The effect of SDF-1 in promoting MSC migration is related to mitogen-activated protein kinase, phosphatidylinositol phospholipase C, and protein kinase signal pathways.</p>


Subject(s)
Animals , Rats , Adenoviridae , Genetics , Cell Movement , Genetics , Physiology , Cells, Cultured , Chemokine CXCL12 , Genetics , Physiology , Enzyme Inhibitors , Pharmacology , Genetic Vectors , Genetics , Mesenchymal Stem Cells , Cell Biology , Metabolism , Mitogen-Activated Protein Kinases , Metabolism , Rats, Wistar , Signal Transduction , Transfection , Type C Phospholipases , Metabolism
13.
Chinese Journal of Cardiology ; (12): 750-756, 2007.
Article in Chinese | WPRIM | ID: wpr-307207

ABSTRACT

<p><b>OBJECTIVE</b>To construct prokaryotic expression vector of pET15b-PEP-1-SOD1 and investigate whether PEP-1-SOD1 fusion protein could be transduced into human umbilical vein endothelial cells (HUVECs) and the effects on hypoxia/reoxygenation injury.</p><p><b>METHODS</b>The recombinant plasmids pET15b-SOD1 and pET15b-PEP-1-SOD1 were constructed and transformed into E. coli BL21 (DE3) to express SOD1 and PEP-1-SOD1 with an N-terminal His-tag. The purified SOD1 and PEP-1-SOD1 were incubated with HUVECs and the viability (MTT assay) and the release of lactate dehydrogenase (LDH) in culture medium were determined in the hypoxia/reoxygenation injury model. The morphological changes were observed under an inverted phase contrast microscope. The content of malondialdehyde (MDA) in HUVECs was also determined with the method of thiobarbituric acid.</p><p><b>RESULTS</b>PEP-1-SOD1 fusion protein could be transduced into cultured HUVECs in a time- and dose-dependent manner. The intracellular enzymatic activity of PEP-1-SOD1 after 30 min incubation with HUVECs was significantly higher than control group (60.88 U/ml +/- 6.73 U/ml vs. 41.06 U/ml +/- 4.19 U/ml, P < 0.01). The transduced PEP-1-SOD1 protein was enzymatically stable for 24 h within cells. After hypoxia/reoxygenation injury, control HUVECs shrunk, became round-shaped and intercellular space increased, while these morphological changes were not observed in PEP-1-SOD1 transduced HUVECs. PEP-1-SOD1 transduction also markedly increased the viability, decreased LDH leakage into culture media and reduced the content of MDA post hypoxia/reoxygenation.</p><p><b>CONCLUSIONS</b>PEP-1-SOD1 fusion protein could be efficiently transduced into HUVECs in a natively active form, and the delivered enzymatically active PEP-1-SOD1 exhibits cellular protection against hypoxia/reoxygenation injury in HUVECs. The transduction of SOD1 mediated by cell-penetrating peptide, PEP-1, provides a basis for further research on the prevention of ischemia/reperfusion injury in vivo.</p>


Subject(s)
Humans , Cell Hypoxia , Cells, Cultured , Cysteamine , Metabolism , Endothelial Cells , Cell Biology , Malondialdehyde , Metabolism , Peptides , Genetics , Metabolism , Recombinant Fusion Proteins , Genetics , Metabolism , Reperfusion Injury , Superoxide Dismutase , Genetics , Metabolism , Transduction, Genetic , Umbilical Veins , Cell Biology
14.
Acta Academiae Medicinae Sinicae ; (6): 93-97, 2007.
Article in Chinese | WPRIM | ID: wpr-230025

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the penetrating ability of fusion protein PEP-1-EGFP with human umbilical vein endothelial cells.</p><p><b>METHODS</b>Two prokaryotic expression plasmids pET15b-EGFP and pET15b-PEP-1-EGFP were constructed and transformed into E. coli BL21 (DE3) to express EGFP and fusion protein PEP-1-EGFP, respectively. The expressed EGFP and PEP-1-EGFP were purified with Ni(2+) -resin affinity chromatography, and their capabilities of transduction into human umbilical vein endothelial cells were evaluated. The time- and dose-dependent transduction of the fusion protein PEP-1-EGFP and its stability in the human umbilical vein endothelial cells were observed. The toxicity of the fusion protein PEP-1-EGFP was detected by MTT method.</p><p><b>RESULTS</b>EGFP failed to be transduced into human umbilical vein endothelial cells, whereas PEP-1-EGFP fusion protein was transduced into cells shortly in 5 minutes. Its transduction was time- and dose-dependent and the fluorescence in the cells were detected even 27 hours later. No cytotoxicity of the fusion protein PEP-1-EGFP to human umbilical vein endothelial cells was detected even when the dose reached up to 200 micromol/L.</p><p><b>CONCLUSION</b>PEP-1-EGFP fusion protein can efficiently transduce the target protein into human umbilical vein endothelial cells, which provides a basis for future researches on the transduction of antioxidant enzymes mediated by the cell-penetrating peptide, PEP-1, in ischemia-reperfusion injury therapy.</p>


Subject(s)
Humans , Cells, Cultured , Cysteamine , Metabolism , Endothelial Cells , Metabolism , Green Fluorescent Proteins , Metabolism , Peptides , Metabolism , Protein Transport , Recombinant Fusion Proteins , Metabolism , Toxicity , Umbilical Veins , Cell Biology
15.
West China Journal of Stomatology ; (6): 166-172, 2007.
Article in Chinese | WPRIM | ID: wpr-348073

ABSTRACT

<p><b>OBJECTIVE</b>To compare the difference in strength degradation and morphology damage of two dental ceramic materials after Hertzian contact cyclic fatigue.</p><p><b>METHODS</b>Hertzian contact technique was used to investigate the response of Empress II glass ceramic and GI- II glass-infiltrated alumina ceramic to cyclic fatigue. Critical loads of specimens after different fatigue cycles were recorded.</p><p><b>RESULTS</b>For Empress II glass ceramic, critical load had significantly difference between specimens after 10(5) cycles loading. No significant difference of critical load was found in GI- II glass-infiltrated alumina ceramic after cycles loading.</p><p><b>CONCLUSION</b>GI- II glass-infiltrated alumina ceramic has better capability in resistance to cyclic loading. It may attribute to microstructure of material. Empress II glass ceramic shows a brittle damage model.</p>


Subject(s)
Humans , Aluminum Oxide , Aluminum Silicates , Ceramics , Dental Porcelain , Glass , Materials Testing
16.
Chinese Journal of Cardiology ; (12): 932-938, 2006.
Article in Chinese | WPRIM | ID: wpr-238488

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the transduction ability of PEP-1-CAT fusion protein into human umbilical vein endothelial cell (HUVECs) and the effects on hydrogen-peroxide (H2O2)-induced oxidative stress injury in these cells.</p><p><b>METHODS</b>With the use of TA-cloning program and isocaudamer technique, the pET15b-PEP-1-CAT of prokaryotic expression plasmid was successfully constructed. The recombinant plasmid was transformed into E.coli BL21 (DE3) and the protein expression was induced by IPTG. The recombinant protein has an N-terminal His-tag which could be used to purify the target protein by affinity chromatography on a Ni2+-NTA-resin column. The fusion protein PEP-1-CAT was prepared and confirmed by specific enzyme activity in vitro. The purified PEP-1-CAT fusion protein was added on cultured HUVECs in vitro. The transduction ability of PEP-1-CAT fusion protein into cells was analyzed by Western blot and specific enzyme activity. The cells were treated with H2O2 (0.5 mmol/L) alone and in combination with PEP-1-CAT fusion protein for 4 h. Then, the cell viability, lactate dehydrogenase (LDH) and malondialdehyde (MDA) contents were measured.</p><p><b>RESULTS</b>The PEP-1-CAT fusion protein could be transduced into the cultured HUVECs in a dose- and time-dependent manner and be stable for at least 48 h. After H2O2 administration, cell viability was significantly reduced compared with control group (37.23%+/-5.68% vs. 100%, P<0.05), while LDH leakage (849.3 U/L+/-95.1 U/L) and MDA (8.23 nmol/L+/-1.58 nmol/L) content were significantly higher than that in control group (540.6 U/L+/-65.7 U/L and 2.46 nmol/L+/-1.42 nmol/L, respectively, all P<0.05). Preincubation with PEP-1-CAT proteins at various concentrations (0.25-2 micromol/L) significantly attenuated H2O2-induced cell injury.</p><p><b>CONCLUSION</b>The PEP-1-CAT fusion protein could efficiently penetrate HUVECs and the transduced protein could attenuate cellular oxidative stress injury induced by H2O2. The PEP-1-CAT fusion protein might be a new strategy for preventing and treating oxidative stress induced diseases.</p>


Subject(s)
Humans , Catalase , Metabolism , Cells, Cultured , Cysteamine , Metabolism , Endothelial Cells , Metabolism , Hydrogen Peroxide , Oxidative Stress , Physiology , Peptides , Metabolism , Umbilical Veins , Cell Biology
17.
Journal of Southern Medical University ; (12): 1114-1117, 2006.
Article in Chinese | WPRIM | ID: wpr-334983

ABSTRACT

<p><b>OBJECTIVE</b>To construct the expression vector pET15b-pep-1-EGFP and purify the fusion protein PEP-1-EGFP expressed in E. coli BL21(DE(3)) for evaluating the cell-penetrating capability of the cell-penetrating peptide PEP-1.</p><p><b>METHODS</b>Two oligonucleotides encoding PEP-1 was synthesized and annealed to generate PEP-1-encoding DNA. The recombinant plasmid pET15b-pep-1-EGFP was constructed by inserting PEP-1-encoding DNA and enhanced green fluorescent protein (EGFP) cDNA into pET15b. The fusion protein PEP-1-EGFP expressed in E. coli BL21(DE(3)) was purified with Ni(2+)-resin affinity chromatography and transduced into human umbilical vein endothelial cells.</p><p><b>RESULTS</b>Sequence analysis confirmed successful construction of the expression vector pET15b-pep-1-EGFP, and the fusion protein PEP-1-EGFP was expressed and purified efficiently with a yield of approximately 14.15 mg/100 ml bacteria medium. SDS-PAGE and Western blotting identified the purified protein as PEP-1-EGFP, and the cell-penetration assay verified that the fusion protein could be transduced into human umbilical vein endothelial cells.</p><p><b>CONCLUSION</b>The successful expression and purification of PEP-1-EGFP and its efficient transduction into human umbilical vein endothelial cells provides a basis for PEP-1-mediated biomacromolecular transduction in protein therapy.</p>


Subject(s)
Humans , Base Sequence , Blotting, Western , Cells, Cultured , Cysteamine , Metabolism , Electrophoresis, Polyacrylamide Gel , Endothelial Cells , Cell Biology , Metabolism , Green Fluorescent Proteins , Genetics , Metabolism , Microscopy, Fluorescence , Molecular Sequence Data , Peptides , Genetics , Metabolism , Plasmids , Genetics , Recombinant Fusion Proteins , Genetics , Metabolism , Transfection , Umbilical Veins , Cell Biology
18.
Journal of Southern Medical University ; (12): 1319-1325, 2006.
Article in Chinese | WPRIM | ID: wpr-334932

ABSTRACT

<p><b>OBJECTIVE</b>To construct the prokaryotic expression plasmid pET15b-PEP-1-CAT to obtain purified fusion protein of PEP-1-CAT.</p><p><b>METHODS</b>Using pfu DNA polymerase, the full-length human catalase cDNA was amplified by PCR from pZeoSV2(+)-CAT plasmid, and the PCR product was added with "A" using Taq DNA polymerase. The purified product of CAT cDNA with the base A at its 3' end was ligated with pGEM-T Easy vector and transformed into DH5alpha. The correct recombinant was identified by PCR and Sal I/Bgl II digestion and named as pGEM-T-CAT. Two oligonucleotides were synthesized and annealed to generate a double-stranded oligonucleotide encoding the PEP-1 peptide, which was directly ligated into Nde I/Xho I-digested pET15b. The recombinant plasmid was identified by double-enzyme digestion and named as pET15b-PEP-1. pET15b-PEP-1 and pGEM-T-CAT were further digested by Xho I/BamH I and Sal I/Bgl II, respectively. The purified linear fragment of pET15b-PEP-1 and CAT cDNA fragment were ligated using two pairs of isocaudarners possessing different recognition sequences but producing compatible cohesive ends. The clone with the expected insert was selected using Xho I restriction analysis followed by sequence analysis. The recombinant plasmid was transformed into E. coli BL21(DE3) which was induced by IPTG. The recombinant protein possessed an N-terminal His-tag sequence which could be used to purify the target protein by affinity chromatography on a Ni(2+)-NTA-resin column. The fusion protein PEP-1-CAT was produced and confirmed by specific enzyme activity in vitro.</p><p><b>RESULTS</b>Sequence analysis showed that the PEP-1 and the human CAT cDNA sequence of pET15b- PEP-1-CAT had identical sequence with designed PEP-1 peptide and human catalase cDNA sequence in GenBank (accession No. AY028632), respectively. SDS-PAGE and Western blotting confirmed successful expression and purification of PEP-1-CAT fusion protein with specific activity of 77.15 U/g.</p><p><b>CONCLUSION</b>The prokaryotic expression plasmid pET15b-PEP-1-CAT has been constructed successfully, and the successful expression and purification of PEP-1-CAT provides a basis for prevention and therapy of various disorders related to oxidative stress.</p>


Subject(s)
Humans , Base Sequence , Blotting, Western , Catalase , Genetics , Metabolism , Chromatography, Affinity , Cloning, Molecular , Cysteamine , Metabolism , Electrophoresis, Polyacrylamide Gel , Escherichia coli , Genetics , Metabolism , Gene Expression , Molecular Sequence Data , Peptides , Genetics , Metabolism , Plasmids , Genetics , Prokaryotic Cells , Metabolism , Recombinant Fusion Proteins , Genetics , Metabolism
SELECTION OF CITATIONS
SEARCH DETAIL