Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Chinese Medical Journal ; (24): 1061-1068, 2011.
Article in English | WPRIM | ID: wpr-239893

ABSTRACT

<p><b>BACKGROUND</b>The high mobility group A1 (HMGA1) proteins are architectural transcription factors found to be overexpressed in lung adenocarcinoma. Lentivirus-mediated RNA interference (RNAi) technology is a powerful tool for silencing endogenous or exogenous genes in human cancer cells. Our preliminary study shows that gemcitabine inhibits growth of the human lung cancer cell line SPCA-1 and induces apoptosis, and this effect might link with down-regulation of HMGA1 expression. This study aimed to investigate the chemosensitivity change of the lung adenocarcinoma cells SPCA-1 after HMGA1 inhibition by lentivirus-mediated RNAi.</p><p><b>METHODS</b>We studied a highly malignant lung adenocarcinoma cell line (SPCA-1 cells). Lentiviral short-hairpin RNA (shHMGA1) expression vectors targeting HMGA1 were used for generation of lentiviral particles. After being transfected into the lung adenocarcinoma cell line SPCA-1, the expression of HMGA1 was determined by retrotranscriptase polymerase chain reaction (RT-PCR) and Western blotting. The effect of gemcitabine on proliferation of positive and negative cells was observed by methyl thiazolyl tetrazolium (MTT) assay and clonogenic survival assay. Apoptosis was observed by flow cytometery. Chemosensitivity to gemcitabine was determined by IC50 analysis. Caspase activity was quantitated by a caspase colorimetric protease assay kit.</p><p><b>RESULTS</b>HMGA1-siRNA silenced its target mRNA specifically and effectively in SPCA-1 cells. The apoptotic rates of the scramble control group were (7.43 ± 0.21)%, (11.00 ± 0.20)%, and (14.93 ± 0.31)%, and the apoptotic rates in the silenced group were (9.53 ± 0.42)%, (16.67 ± 0.45)%, and (25.40 ± 0.79)% under exposure to 0.05, 0.5 and 5.0 µg/ml of gemcitabine (P < 0.05). The IC(50) of the silenced group was (0.309 ± 0.003) µg/ml which was significantly lower than in the scramble control group, (0.653 ± 0.003) µg/ml (P < 0.05). It reduced cancer cell proliferation and increased apoptotic cell death after being treated with gemcitabine compared with the scramble control group. HMGA1 silencing resulted in reduction in the phosphorylation of Akt, and promoted the activation of caspases 3, 8 and 9 upon exposure to gemcitabine.</p><p><b>CONCLUSIONS</b>Lentivirus-mediated RNA interference of HMGA1 enhanced chemosensitivity to gemcitabine in lung adenocarcinoma cells. The mechanism may be associated with the PI-3K/Akt signal pathway. HMGA1 may represent a novel therapeutic target in lung cancer.</p>


Subject(s)
Humans , Antimetabolites, Antineoplastic , Pharmacology , Blotting, Western , Calcium-Transporting ATPases , Genetics , Metabolism , Caspase 3 , Genetics , Metabolism , Caspase 8 , Genetics , Metabolism , Caspase 9 , Genetics , Metabolism , Cell Line, Tumor , Cell Proliferation , Deoxycytidine , Pharmacology , Flow Cytometry , Genetic Vectors , Genetics , HMGA Proteins , Genetics , Metabolism , Lentivirus , Genetics , RNA Interference , Physiology , Reverse Transcriptase Polymerase Chain Reaction
2.
Journal of Experimental Hematology ; (6): 67-73, 2010.
Article in English | WPRIM | ID: wpr-328570

ABSTRACT

This study was aimed to explore the potential therapy of Gambogic acid (GA) combined with magnetic nanoparticle of Fe3O4 (Fe3O4-MNP) on leukemia. The proliferation of U937 cells and the cytotoxicity were evaluated by MTT assay. Cell apoptosis was observed and analyzed by microscopy and flow cytometry respectively. The expressions of gene and protein were detected by quantitative real-time polymerase chain reaction and Western blot respectively. The results showed that GA enhanced the cytotoxicity for U937 cells in dose- and time-dependent manners. The Fe3O4-MNP itself had not cytotoxicity, but could enhance the inhibitory effect of GA on proliferation of U937 cells. The apoptotic rate of U937 cells induced by combination of GA with Fe3O4-MNP was higher than that by GA alone. The typical apoptotic features of cells treated with GA and Fe3O4-MNP were observed. The expression levels of caspase-3 and bax after co-treatment of GA and Fe3O4-MNP were higher than that exposed to GA or Fe3O4-MNP alone, but the expressions of bcl-2, NF-kappaB and survivin were down-regulated. It is concluded that Fe3O4-MNP can promote GA-induced apoptosis in U937 cells, and the combination of GA with Fe3O4-MNP may be a safer and less toxic new therapy for leukemia.


Subject(s)
Humans , Apoptosis , Iron Compounds , Pharmacology , Magnetics , Nanoparticles , U937 Cells , Xanthones , Pharmacology
3.
Chinese Medical Journal ; (24): 3427-3432, 2010.
Article in English | WPRIM | ID: wpr-336608

ABSTRACT

<p><b>BACKGROUND</b>Platinum-based chemotherapeutics are the most common regimens for advanced non-small-cell lung cancer (NSCLC) patients, and genetic factors are thought to represent important determinants of drug efficacy. We prospectively assessed the status of the XPC Ala499Val and Lys939Gln gene polymorphisms and investigated whether these SNPs can predict the response to cisplatin/carboplatin-based regimens in advanced NSCLC patients in a Chinese population.</p><p><b>METHODS</b>The treatment outcomes of 96 advanced NSCLC patients who were treated with platinum-based chemotherapy were evaluated. The polymorphic status of xeroderma pigmentosum group C (XPC) gene was genotyped by the 3-D polyacrylamide gel-based DNA microarray method.</p><p><b>RESULTS</b>The distributions of XPC Lys939Gln genotypes differed significantly between the response group (complete + partial responses) and the non-response group (stable + progressive disease; P = 0.022). The heterozygous A/C genotype carriers had a poorer response rate than the wild A/A genotype carriers in stage III (OR, 0.074; 95%CI, 0.008 - 0.704; P = 0.023). The XPC Ala499Val polymorphisms were not associated with response to platinum-based chemotherapy.</p><p><b>CONCLUSION</b>Polymorphisms of the XPC gene, Lys939Gln, may be a predictive marker of treatment response for advanced NSCLC patients in stage III.</p>


Subject(s)
Adult , Aged , Female , Humans , Male , Middle Aged , Alleles , Antineoplastic Combined Chemotherapy Protocols , Therapeutic Uses , Carboplatin , Carcinoma, Non-Small-Cell Lung , Drug Therapy , Genetics , Pathology , Cisplatin , DNA-Binding Proteins , Genetics , Genotype , Lung Neoplasms , Drug Therapy , Genetics , Pathology , Neoplasm Staging , Polymorphism, Single Nucleotide , Prospective Studies
4.
Journal of Experimental Hematology ; (6): 54-59, 2009.
Article in English | WPRIM | ID: wpr-302198

ABSTRACT

The aim of this study was to investigate the potential benefit of combination therapy with magnetic nanoparticle of Fe(3)O(4) and 5-Bromotetrandrine (5-BrTet) on chronic leukemia. The apoptosis was detected by flow cytometry (FCM), Wright staining and light microscope; the expressions of BAX and BCL-2 were measured by Western blot. The results showed that combination of daunorubicin (DNR) with either MNP (Fe(3)O(4)) or 5-BrTet exerted a potent cytotoxic effect on K562/A02 cells, while MNP (Fe(3)O(4)) and 5-BrTet co-treatment could synergistically enhance DNR-induced apoptosis. After treated with this regimen, the typical apoptotic morphological features were found in K562/A02 cells; the expression level of BCL-2 decreased and BAX increased markedly. It is concluded that MNP (Fe(3)O(4)) or 5-BrTet with DNR can induce apoptosis in K562/A02 cells, and they show distinct synergism when used together. The down-regulation of BCL-2 and the up-regulation of BAX may play important roles.


Subject(s)
Humans , Apoptosis , Benzylisoquinolines , Pharmacology , Daunorubicin , Pharmacology , Down-Regulation , Ferric Compounds , Gene Expression Regulation, Leukemic , K562 Cells , Nanoparticles , Proto-Oncogene Proteins c-bcl-2 , Metabolism , Up-Regulation , bcl-2-Associated X Protein , Metabolism
5.
Journal of Experimental Hematology ; (6): 60-64, 2009.
Article in English | WPRIM | ID: wpr-302197

ABSTRACT

This study was aimed to investigate the reversal effect of 5-bromotetrandrine (5-BrTet) and magnetic nanoparticle of Fe(3)O(4) (Fe(3)O(4)-MNPs) combined with DNR in vivo. The xenograft leukemia model with stable multiple drug resistance in nude mice was established. The two sub-clones of K562 and K562/A02 cells were respectively inoculated subcutaneously into back of athymic nude mice (1 x 10(7) cells/each) to establish the leukemia xenograft models. Drug resistant and the sensitive tumor-bearing nude mice were both assigned randomly into 5 groups: group A was treated with NS; group B was treated with DNR; group C was treated with nanoparticle of Fe(3)O(4) combined with DNR; group D was treated with 5-BrTet combined with DNR; group E was treated with 5-bromotetrandrine and magnetic nanoparticle of Fe(3)O(4) combined with DNR. The incidence of tumor formation, growth characteristics, weight and volume of tumor were observed. The histopathologic examination of tumors and organs were carried out. The protein levels of BCL-2, BAX, and Caspase-3 in resistant tumors were detected by Western blot. The results indicated that 5-BrTet and magnetic nanoparticle of Fe(3)O(4) combined with DNR significantly suppressed growth of K562/A02 cell xenograft tumor, histopathologic examination of tumors showed the tumors necrosis obviously. Application of 5-BrTet and magnetic nanoparticle of Fe(3)O(4) inhibited the expression of BCL-2 protein and up-regulated the expression of BAX, and Caspase-3 protein in K562/A02 cell xenograft tumor. It is concluded that 5-bromotetrandrine and magnetic nanoparticle of Fe(3)O(4) combined with DNR have significant tumor-suppressing effect on MDR leukemia cell xenograft model.


Subject(s)
Animals , Female , Humans , Mice , Antineoplastic Agents , Pharmacology , Benzylisoquinolines , Pharmacology , Daunorubicin , Pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Synergism , Ferric Compounds , K562 Cells , Mice, Inbred BALB C , Mice, Nude , Nanoparticles , Xenograft Model Antitumor Assays
6.
Journal of Experimental Hematology ; (6): 345-351, 2009.
Article in English | WPRIM | ID: wpr-302135

ABSTRACT

Multidrug resistance (MDR) plays a major role in the failure of cancer chemotherapy. Since Fe(3)O(4)-magnetic nanoparticle loaded with daunorubicin (DNR) can overcome multidrug-resistance of K562 cells in vitro, the effect of Fe(3)O(4)-magnetic nanoparticle loaded with DNR on multidrug-resistant K562 cells was studied in vivo, the K562-n and its MDR counterpart K562-n/VCR cells were inoculated subcutaneously into both sides of the back of nude mice to establish a human leukemia xenograft model. The mice were randomly divided into group A receiving normal saline, group B receiving DNR, group C receiving Fe(3)O(4)-magnetic nanoparticle, group D receiving Fe(3)O(4)-magnetic nanoparticle loaded with DNR and group E receiving Fe(3)O(4)-magnetic nanoparticle containing DNR with a magnetic field built on the surface of the tumor tissue. The tumor volume was measured on the day 1, 5, 9, 13, 17 and 21 after the first treatment. Tumor tissues were isolated for examination of the expression of mdr-1 by reverse transcription polymerase chain reaction and Western blotting. The results showed that for K562-n/VCR tumor, the tumor volume was markedly lower in groups D and E than that in groups A, B and C. Pathological observation revealed that the tumor cells of group A and B grew well, some disseminated necrosis and some cells with karyorrhexis and karyopyknosis existed in group C. However, significant fracture, necrosis of cell and subsequently fibrosis were seen in group D and E. The transcription of mdr-1 gene in groups D and E was significantly lower than that in groups A, B and C (group D and E vs group A, B or C, p < 0.05). However, there were no differences about the protein expression of P-gp between these groups. The tumor volume of K562-n in groups C, D and E was markedly lower than that in groups A and B (group C, D and E vs group A or B, p < 0.05). Pathological observation showed that the tumor cell of group A and B grew well, and no obvious necrosis was observed. Significant fracture, necrosis of cell and subsequently fibrosis were seen in group C, D and E. It is concluded that DNR-loaded Fe(3)O(4) magnetic nanoparticles can suppress the growth of the MDR K562-n/VCR tumor in vivo, but can not further enhance its efficacy on the sensitive K562-n tumor as compared to DNR alone. The additional external magnetic field failed to further improve the antitumor effect in vivo.


Subject(s)
Animals , Female , Humans , Mice , Daunorubicin , Pharmacology , Therapeutic Uses , Drug Carriers , Drug Resistance, Multiple , Drug Resistance, Neoplasm , K562 Cells , Leukemia , Drug Therapy , Magnetics , Mice, Inbred BALB C , Mice, Nude , Nanoparticles , Treatment Outcome , Xenograft Model Antitumor Assays
7.
Journal of Experimental Hematology ; (6): 1179-1182, 2009.
Article in Chinese | WPRIM | ID: wpr-343323

ABSTRACT

This study was aimed to investigate the reversal effect of Tetrandrine (TET) combined with daunorubicin (DNR) on multidrug resistance (MDR) of K562/A02 cells and its relation to P21, P-gp and their genes so as to provide the new theoretic evidence for clinical use of TET. The experiments were divided into 4 groups: control group (DNR alone), combined 1 group (DNA+0.5 mg/L TET), combined 2 group (DNR+1.0 mg/L TET) and combined 3 group (DNR+2.0 mg/L TET). The expressions of P21, P-gp and mdr-1 gene in K562/A02 cells of different groups were detected by Western blot, flow cytometry and semi-quantitative PCR respectively. The results showed that the expression of P21 was enhanced along with increasing of TET concentration, the expression of P-gp was reduced along with increasing of TET concentration and expression of mdr-1 gene was almost not observed in K562 cells, but the high expression of mdr-1 gene was seen in K562/A02 cells, furthermore, the expression of mdr-1 gene in K562/A02 cells increasingly was reduced along with increasing of TET concentration. It is concluded that the TET possesses the reversal effect on multiple drug resistance of K562/A02 cells with concentration dependence, the reversal effect of TET may be related to up-regulation of P21 expression and down-regulation of P-gp and mdr-1 gene expressions in K562/A02 cells.


Subject(s)
Humans , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Metabolism , Benzylisoquinolines , Pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21 , Metabolism , Daunorubicin , Pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , K562 Cells
8.
Journal of Experimental Hematology ; (6): 1183-1191, 2009.
Article in English | WPRIM | ID: wpr-343322

ABSTRACT

The present study was aimed to evaluate the MDR reversal activity of bromotetrandrine (BrTet) in vitro and in vivo. The inhibitory effects of adriamycin (ADM) used alone or in combination with BrTet or Tet on the proliferation of K562 and K562/A02 cells were evaluated by MTT assay. The ADM accumulation and the protein levels of P-glycoprotein (P-gp) were detected by flow cytometry. The mRNA levels of P-gp were determined by RT-PCR. The in vivo effect of BrTet and Tet was investigated by using nude mice grafted with sensitive human leukemia cell line K562 and MDR cell line K562/A02. The results showed that BrTet at 0.25, 0.5 and 1 micromol/L reversed the resistance to ADM in MDR K562/A02 cells in a dose-dependent manner. Flow cytometry suggested that BrTet significantly increased the intracellular accumulation of ADM in K562/A02 cells in a dose-dependent manner. BrTet also inhibited the overexpression of P-gp in K562/A02 cells, and down-regulated mdr1 expression. In nude mice bearing K562 xenografts on the left flank and K562/A02 xenografts on the right flank, intraperitoneal injection of 10 mg/kg BrTet significantly enhanced the antitumor activity of ADM against K562/A02 xenografts with inhibitory rates of 26.1%, while ADM alone inhibited the growth of K562/A02 xenografts only by 5.8%. No enhancement effect by BrTet was seen in K562 xenografts. It is concluded that BrTet shows significant MDR reversal activity in vitro and in vivo. Its activity may be related to the inhibition of P-gp overexpression and the increase intracellular accumulation of anticancer drugs. BrTet may be a promising-MDR modulator for eventual assessment in the clinic.


Subject(s)
Animals , Female , Humans , Mice , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Metabolism , Benzylisoquinolines , Pharmacology , Drug Resistance, Multiple , Genetics , Drug Resistance, Neoplasm , Genetics , K562 Cells , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
9.
Journal of Experimental Hematology ; (6): 1283-1287, 2008.
Article in Chinese | WPRIM | ID: wpr-234249

ABSTRACT

This study was aimed to investigate the effects of low frequency and power ultrasound combined with adriamycin on apoptosis of drug-resistant leukemia cell line K562/A02 in vitro, to find out the parameters of optimal exposure, and to explore the possible mechanism reversing drug-resistance of K562/A02 cells. The K562/A02 cells in logarithmic growth phase were used in experiments. The experiments were divided into 4 groups: group control, group adriamycin (A02) alone, group ultrasound (US) alone and group A02+US. The trypan blue dye exclusion test and MTT assay were used to determine the cell viability; Wright's staining was used to detect the apoptosis; the flow cytometry was used to analyze the drug concentration, and the scanning electron microscopy was used to observe the changes of cell surface. The results showed that the significant differences in cell viability, intracellular adriamycin concentration and changes of cell membrane were found between ultrasound-treated and untreated cells in the presence of various concentration of adriamycin. The exposure to ultrasound at 20 kHZ, 0.25 W/cm2 for 60 seconds could obviously decrease LC50 of adriamycin to K562/A02 cells, while the exposure to ultrasound at 20 kHZ, 0.05 W/cm2 for 60 seconds could kill K562/A02 cells at once. After being treated by low frequency ultrasound, the small holes with diameter about 1-2 microm in the cell surface appeared. The ultrasound increased the adriamycin concentration in the cells, accelerated the formation of apoptotic bodies, and promoted apoptosis of adriamycin-resistant cells. It is concluded that the ultrasound at optimal parameters enhances inhibitory effect of adriamycin on drug-resistant cell line, thereby reverses drug-resistance of drug-resistant cell line through sound-hole effect in tumor cells resulting from ultrasound induced cavitation.


Subject(s)
Humans , Doxorubicin , Pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , K562 Cells , Ultrasonics
10.
Chinese Journal of Hematology ; (12): 321-324, 2008.
Article in Chinese | WPRIM | ID: wpr-240017

ABSTRACT

<p><b>OBJECTIVE</b>To study the effect of tetrandrine (Tet) in combination with droloxifen (DRL) on the expression of nuclear factor kappa B (NF-kappaB) in K562 and K562/A02 cell lines and its reversal mechanism.</p><p><b>METHODS</b>The activation of NF-kappaB in K562 and K562/A02 cell lines and the effect of Tet or DRL alone or in combination on NF-kappaB protein expression were determined with immunocytochemistry and Western blotting respectively.</p><p><b>RESULTS</b>(1) K562/A02 cells displayed higher level of NF-kappaB protein expression than K562 cells. (2) The application of Tet or DRL alone or in combination had no effect on NF-kappaB expression in K562 cells at 6 h and 12 h (P > 0.05). (3) Tet and DRL alone or in combination could significantly down-regulate NF-kappaB protein expression in nuclei of K562/A02 cells. The effect was more significant in combination than either alone. This effect was more significant at 12 h than at 6 h.</p><p><b>CONCLUSIONS</b>(1) Activation of NF-kappaB may be involved in the mechanism of MDR of K562/A02 cell line. (2)Inhibition of NF-kappaB activation may be involved in the reversal of multidrug resistance in K562/A02 cells by Tet and DRL.</p>


Subject(s)
Humans , Benzylisoquinolines , Pharmacology , Drug Resistance, Multiple , K562 Cells , NF-kappa B , Metabolism , Tamoxifen , Pharmacology
11.
Biomedical and Environmental Sciences ; (12): 495-501, 2007.
Article in English | WPRIM | ID: wpr-249819

ABSTRACT

<p><b>OBJECTIVE</b>To assess the ability of tetrandrine (Tet) to enhance the sensitivity to irradiation and its mechanism in cell lines of human breast cancer p53-mutant MCF-7/ADR, p53-wild-type MCF-7 and human colon carcinoma p53-mutant HT-29 as well as in C26 colorectal carcinoma-bearing BALB/c mice.</p><p><b>METHODS</b>MCF-7/ADR, HT-29 and MCF-7 cells were exposed to irradiation in the absence or presence of tetrandrine. The effect of Tet on the cytotoxicity of X-irradiation in these three cells was determined and the effect of tetrandrine on cell cycle arrest induced by irradiation in its absence or presence was studied by flow cytometry. Moreover, mitotic index measurement determined mitosis of cells to enter mitosis. Western blotting was employed to detect cyclin B 1 and Cdc2 proteins in extracts from irradiated or non-irradiated cells of MCF-7/ADR, HT-29 and MCF-7 treated with tetrandrine at various concentrations. Tumor growth delay assay was conducted to determine the radio-sensitization of tetrandrine in vivo.</p><p><b>RESULTS</b>Clonogenic assay showed that tetrandrine markedly enhanced the lethal effect of X-rays on p53-mutant MCF-7/ADR and HT-29 cells and the sensitization enhancement ratio (SER) of tetrandrine was 1.51 and 1.63, but its SER was only 1.1 in p53-wt MCF-7 cells. Irradiated p53-mutant MCF-7/ADR and HT-29 cells were only arrested in G2/M phase while MCF-7 cells were arrested in G1 and G2/M phases. Radiation-induced G2 phase arrests were abrogated by tetrandrine in a concentration-dependent manner in MCF-7/ADR and HT-29 cells, whereas redistribution within MCF-7 cell cycle changed slightly. The proportion of cells in M phase increased from 1.3% to 14.7% in MCF-7/ADR cells, and from 1.5% to 13.2% in HT-29 cells, but 2.4% to 7.1% in MCF-7 cells. Furthermore, the levels of cyclin B 1 and Cdc2 expression decreased after X-irradiation in MCF-7/ADR and HT-29 cells, and the mitotic index was also lower. Tet could reverse the decrease and induce the irradiated cells to enter mitosis (M phase). Endosomatic experiment showed that tetrandrine caused tumor growth delay in irradiated mice.</p><p><b>CONCLUSION</b>Tetrandrine boosts the cell killing activity of irradiation both in vitro and in vivo. Tetrandrine is a potent abrogator for G2 checkpoint control and can sensitize the cells to radiation.</p>


Subject(s)
Animals , Humans , Male , Mice , Benzylisoquinolines , Pharmacology , CDC2-CDC28 Kinases , Metabolism , Cell Line, Tumor , Cyclin B , Metabolism , Cyclin B1 , Drug Screening Assays, Antitumor , G2 Phase , Mice, Inbred BALB C , Radiation Tolerance
12.
Acta Pharmaceutica Sinica ; (12): 419-423, 2002.
Article in Chinese | WPRIM | ID: wpr-274854

ABSTRACT

<p><b>AIM</b>To investigate the radiosensitizing effect and mechanism of action of staurosporine (STP) in human colon carcinoma HT-29 and breast cancer MCF-7/ADR cells.</p><p><b>METHODS</b>The effect of STP on the cytotoxicity of X-ray was determined by clonogenic assay. The effect of STP on cell cycle arrest induced by X irradiation was studied in two cell lines by using flow cytometry, Western Blotting was performed to indicate the changes of cyclin B1 and cdc2 protein levels.</p><p><b>RESULTS</b>STP sensitized the two cell lines to X-ray by clonogenic assay. STP potentiated the cytotoxicity of X-ray by 2.10- and 2.09-fold in HT-29 and MCF-7/ADR cells. Flow cytometry assay showed that exposure of HT-29 and MCF-7/ADR cells to X-ray caused cells arrest in G2 phase. The percentage of arrest G2 phase cells were 56% and 52.7%, respectively. The addition of STP after irradiation resulted in a dose-dependent reduction of G2 phase arrest induced by X-ray. Furthermore, the results showed that STP blocked decrease of cyclin B1 expression induced by X-ray, while mitotic index measurement indicated that X-ray-irradiated cells treated with STP entered mitosis. The data suggested that the potentiation of cytotoxicity of X-ray by STP is associated with the suppression of cyclin B1 expression, which result in the abrogation of G2 arrest, before the cells entered into M phase, they had not enough time to repair.</p><p><b>CONCLUSION</b>STP is a potent G2 checkpoint abrogator and markedly enhanced the cytotoxicity of X irradiation in the p53 mutant cancer cells.</p>


Subject(s)
Female , Humans , Breast Neoplasms , Pathology , Cyclin B , Cyclin B1 , Enzyme Inhibitors , Pharmacology , G2 Phase , HT29 Cells , Mitotic Index , Particle Accelerators , Radiation Tolerance , Radiation-Sensitizing Agents , Pharmacology , Staurosporine , Pharmacology , Tumor Cells, Cultured
13.
Chinese Journal of Radiation Oncology ; (6)1992.
Article in Chinese | WPRIM | ID: wpr-678156

ABSTRACT

Objective To investigate the mechanism of the inhibitory effect of ?-ray irradiation on rat vascular smooth muscle cells(VSMC). Methods The ef fect of ?-ray irradiation on proliferation of VSMC was observed by 3?H-TdR incor poration. After ?-ray irradiation, the VSMC cell cycle change was detected b y flo w cytometry. The expression of p53, cyclin D and PCNA was investigated by Wester n Blot. Results The inhibitory effect of ?-ray irradiation on VSMC proliferati on was dose-dependent. After ?-ray irradiation, VSMC was arrested in G 1 st age, w ith the expression of p53 increased but the expression of cyclin D and PCNA decr eased. Conclusions ?-ray irradiation can inhibit the proliferation of VSMC. T he main mechanism is probably due to the induction of cell cycle arrest and inhi bition of the VSMC mitosis ,during which process, p53,cyclin D and PCNA all pla y an important role .

SELECTION OF CITATIONS
SEARCH DETAIL