1
|
Sheremet YA, Yemets AI, Azmi A, Vissenberg K, Verbelen JP, Blume YB. Effects of tyrosine kinase and phosphatase inhibitors on mitosis progression in synchronized tobacco BY-2 cells. CYTOL GENET+ 2012. [DOI: 10.3103/s0095452712050088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
2
|
Li W, Frame LT, Hirsch S, Cobos E. Genistein and hematological malignancies. Cancer Lett 2010; 296:1-8. [DOI: 10.1016/j.canlet.2010.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Revised: 04/29/2010] [Accepted: 05/03/2010] [Indexed: 02/04/2023]
|
3
|
Sasamura H, Takahashi A, Yuan J, Kitamura H, Masumori N, Miyao N, Itoh N, Tsukamoto T. Antiproliferative and antiangiogenic activities of genistein in human renal cell carcinoma. Urology 2005; 64:389-93. [PMID: 15302513 DOI: 10.1016/j.urology.2004.03.045] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2003] [Accepted: 03/29/2004] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To determine whether genistein, an isoflavone that is plentiful in soy beans, could inhibit the growth of human renal cell carcinoma (RCC) cells in vitro, induce apoptosis, and suppress neovascularization in vivo induced by human RCC cells. METHODS We investigated the effect of genistein on cell proliferation in four human RCC cell lines, SMKT R-1, R-2, R-3, and R-4. The terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling assay was performed to examine whether genistein could induce apoptosis in SMKT R-1 cells. To evaluate the effect of genistein on in vivo angiogenesis, we used a new mouse dorsal air sac model in which we could evaluate it simply and quantitatively. Radioisotope-labeled red blood cells were injected into a tail vein in mice bearing a Millipore filter chamber containing genistein, and the vascular volume was examined by measuring the radioactivity of the mouse dorsal skin. RESULTS Treatment with genistein for 48 hours inhibited cell proliferation in a dose-dependent manner and 100 microg/mL genistein inhibited it in a time-dependent manner. A dose of 50 microg/mL genistein clearly induced cell apoptosis. The vascular volume after implantation of the Millipore filter chamber containing RCC cells increased to threefold that without RCC cells. Genistein in the Millipore filter chamber significantly decreased the neovascularization induced by human RCC cells in vivo. CONCLUSIONS The results of this study demonstrated that genistein inhibited cell proliferation, induced apoptosis, and suppressed in vivo angiogenesis in human RCC cells. Genistein may be a promising antitumorigenic and antiangiogenic agent for the treatment and prevention of RCC.
Collapse
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/therapeutic use
- Apoptosis/drug effects
- Carcinoma, Renal Cell/blood supply
- Carcinoma, Renal Cell/pathology
- Cell Division/drug effects
- Cell Line, Tumor/drug effects
- Cell Line, Tumor/physiology
- Diffusion Chambers, Culture
- Drug Screening Assays, Antitumor
- Female
- Genistein/pharmacology
- Genistein/therapeutic use
- Humans
- Kidney Neoplasms/pathology
- Mice
- Mice, Inbred C57BL
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/etiology
Collapse
Affiliation(s)
- Hiroto Sasamura
- Department of Urology, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Nozawa F, Itami A, Saruc M, Kim M, Standop J, Picha KS, Cowan KH, Pour PM. The combination of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/Apo2L) and Genistein is effective in inhibiting pancreatic cancer growth. Pancreas 2004; 29:45-52. [PMID: 15211111 DOI: 10.1097/00006676-200407000-00055] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Our previous studies have shown that, contrary to many other human pancreatic adenocarcinoma cell lines, AsPC1 cells are resistant to the apoptotic effect of the tumor necrosis factor-related apoptosis-inducing ligand, also called Apo2L (TRAIL/Apo2L). In our in vitro studies, the combination of TRAIL/Apo2L and protein synthesis inhibitor, genistein, but not genistein alone, was, however, effective in inducing apoptosis in AsPC1 cells. In the present study, we examined the effect of TRAIL/Apo2L with genistein on the growth of AsPC1 cells in vitro and in vivo. METHODS Mice with orthotopically transplanted AsPC1 cells were treated either with TRAIL/Apo2L, Genistein (Gen) or a combination of both (TRAIL/Apo2L + Gen) for 14 days. After 14 days, the size and weight of the tumors were registered and the apoptosis of the tumor cells were determined by the TUNEL method. In vitro, the effect of combination treatment on cytotoxicity was assessed by MTT assay and apoptosis was assessed by DAPI staining. FADD, caspase 3, and PARP proteins were determined by Western blot. RESULTS No toxic side effects were observed in either group. The tumor volume was significantly smaller and the apoptotic ratio was higher in the TRAIL + Gen group than in the other 2 groups. The apoptotic effect was associated with the caspase-3 activation. Z-VAD-FMK partially inhibited apoptosis by TRAIL + Gen. CONCLUSIONS These results indicate that the combination of TRAIL/Apo2L with genistein presents a promising therapeutic approach for the treatment of pancreatic cancer. Further detail investigations are needed, however, to verify the mechanisms of this combination therapy.
Collapse
Affiliation(s)
- Fumiaki Nozawa
- UNMC Eppley Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198-6805, USA
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Ravindranath MH, Muthugounder S, Presser N, Viswanathan S. Anticancer therapeutic potential of soy isoflavone, genistein. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2004; 546:121-65. [PMID: 15584372 DOI: 10.1007/978-1-4757-4820-8_11] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Genistein (4'5, 7-trihydroxyisoflavone) occurs as a glycoside (genistin) in the plant family Leguminosae, which includes the soybean (Glycine max). A significant correlation between the serum/plasma level of genistein and the incidence of gender-based cancers in Asian, European and American populations suggests that genistein may reduce the risk of tumor formation. Other evidence includes the mechanism of action of genistein in normal and cancer cells. Genistein inhibits protein tyrosine kinase (PTK), which is involved in phosphorylation of tyrosyl residues of membrane-bound receptors leading to signal transduction, and it inhibits topoisomerase II, which participates in DNA replication, transcription and repair. By blocking the activities of PTK, topoisomerase II and matrix metalloprotein (MMP9) and by down-regulating the expression of about 11 genes, including that of vascular endothelial growth factor (VEGF), genistein can arrest cell growth and proliferation, cell cycle at G2/M, invasion and angiogenesis. Furthermore, genistein can alter the expression of gangliosides and other carbohydrate antigens to facilitate their immune recognition. Genistein acts synergistically with drugs such as tamoxifen, cisplatin, 1,3-bis 2-chloroethyl-1-nitrosourea (BCNU), dexamethasone, daunorubicin and tiazofurin, and with bioflavonoid food supplements such as quercetin, green-tea catechins and black-tea thearubigins. Genistein can augment the efficacy of radiation for breast and prostate carcinomas. Because it increases melanin production and tyrosinase activity, genistein can protect melanocytes of the skin of Caucasians from UV-B radiation-induced melanoma. Genistein-induced antigenic alteration has the potential for improving active specific immunotherapy of melanoma and carcinomas. When conjugated to B43 monoclonal antibody, genistein becomes a tool for passive immunotherapy to target B-lineage leukemias that overexpress the target antigen CD19. Genistein is also conjugated to recombinant EGF to target cancers overexpressing the EGF receptor. Although genistein has many potentially therapeutic actions against cancer, its biphasic bioactivity (inhibitory at high concentrations and activating at low concentrations) requires caution in determining therapeutic doses of genistein alone or in combination with chemotherapy, radiation therapy, and/or immunotherapies. Of the more than 4500 genistein studies in peer-reviewed primary publications, almost one fifth pertain to its antitumor capabilities and more than 400 describe its mechanism of action in normal and malignant human and animal cells, animal models, in vitro experiments, or phase I/II clinical trials. Several biotechnological firms in Japan, Australia and in the United States (e.g., Nutrilite) manufacture genistein as a natural supplement under quality controlled and assured conditions.
Collapse
Affiliation(s)
- Mepur H Ravindranath
- Laboratory of Glycoimmunotherapy, John Wayne Cancer Institute, 2200 Santa Monica Blvd., Santa Monica, CA 90404-2302, USA.
| | | | | | | |
Collapse
|
6
|
Conklin KA. Dietary antioxidants during cancer chemotherapy: impact on chemotherapeutic effectiveness and development of side effects. Nutr Cancer 2001; 37:1-18. [PMID: 10965514 DOI: 10.1207/s15327914nc3701_1] [Citation(s) in RCA: 280] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Several studies suggest that dietary supplementation with antioxidants can influence the response to chemotherapy as well as the development of adverse side effects that results from treatment with antineoplastic agents. Administration of antineoplastic agents results in oxidative stress, i.e., the production of free radicals and other reactive oxygen species (ROS). Oxidative stress reduces the rate of cell proliferation, and that occurring during chemotherapy may interfere with the cytotoxic effects of antineoplastic drugs, which depend on rapid proliferation of cancer cells for optimal activity. Antioxidants detoxify ROS and may enhance the anticancer effects of chemotherapy. For some supplements, activities beyond their antioxidant properties, such as inhibition of topoisomerase II or protein tyrosine kinases, may also contribute. ROS cause or contribute to certain side effects that are common to many anticancer drugs, such as gastrointestinal toxicity and mutagenesis. ROS also contribute to side effects that occur only with individual agents, such as doxorubicin-induced cardiotoxicity, cisplatin-induced nephrotoxicity, and bleomycin-induced pulmonary fibrosis. Antioxidants can reduce or prevent many of these side effects, and for some supplements the protective effect results from activities other than their antioxidant properties. Certain side effects, however, such as alopecia and myelosuppression, are not prevented by antioxidants, and agents that interfere with these side effects may also interfere with the anticancer effects of chemotherapy.
Collapse
Affiliation(s)
- K A Conklin
- Department of Anesthesiology, UCLA School of Medicine, Center for the Health Sciences 90095, USA.
| |
Collapse
|
7
|
Drummond MW, Holyoake TL. Tyrosine kinase inhibitors in the treatment of chronic myeloid leukaemia: so far so good? Blood Rev 2001; 15:85-95. [PMID: 11409908 DOI: 10.1054/blre.2001.0152] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Chronic myeloid leukaemia (CML) is characterized by marked expansion of the myeloid series, and is thought to arise as a direct result of the bcr-abl fusion-gene. The BCR-ABL oncoprotein is a constitutively active protein tyrosine kinase (PTK), which results in altered cell signalling and is responsible for the changes that characterize the malignant cells of CML. It has been shown that the increased tyrosine kinase activity of BCR-ABL is a requirement for transformation and is, therefore, a legitimate target for pharmacological inhibition. Several compounds have now been identified as relatively selective inhibitors of BCR-ABL, including members of the tyrphostin family, herbimycin A and most importantly the 2-phenylaminopyrimidine ST1571. Having established the efficacy of this agent in vitro, phase I trials using an oral formulation were commenced in the USA in mid 1998. Early data from an interferon-alpha (IFN) resistant/refractory or intolerant cohort demonstrated good patient tolerance and effective haematological control at doses above 300 mg. More promising was its ability to induce cytogenetic responses in this pretreated group of patients. Phase II data, albeit far from complete, appear to confirm its efficacy even in the context of advanced disease and phase III clinical trials are currently underway in many countries. Recent laboratory evidence, however, suggests that the development of drug resistance is a possibility (via amplification of the bcr-abl fusion gene, overexpression of P-glycoprotein or binding of ST1571 to alpha1 acid glycoprotein) and that combination therapy including ST1571 should be considered.
Collapse
Affiliation(s)
- M W Drummond
- Academic Transfusion Medicine Unit, University Department of Medicine, Glasgow Royal Infirmary, Glasgow, UK.
| | | |
Collapse
|
8
|
Rauko P, Novotny L, Dovinova I, Hunakova L, Szekeres T, Jayaram HN. Antitumor activity of benzamide riboside and its combination with cisplatin and staurosporine. Eur J Pharm Sci 2001; 12:387-94. [PMID: 11231105 DOI: 10.1016/s0928-0987(00)00180-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Benzamide riboside (BR), a new synthetic nucleoside analogue, has demonstrated a potent cytotoxic activity in murine leukemia in vitro. The purpose of the present investigation was to examine the antitumor activity of BR in mice bearing leukemia L1210. The results revealed that BR possesses a potent antitumor activity in vivo. It increases life-span of mice with leukemia. Synergistic cytotoxicity of BR with select DNA damaging agents, cisplatin (cis-Pt) and staurosporine (STP) was examined in MTT chemosensitivity assay, FACS analyses and apoptotic DNA fragmentation on L1210 cells in culture. A simultaneous treatment of leukemia L1210 cells with the combination of BR and STP resulted in synergistic cytotoxicity that correlated with increased apoptotic activity in those cells. On the other hand, treatment of L1210 cells with combination of BR and cis-Pt resulted in antagonistic cytotoxic effect. Finally, to elucidate the synergistic effect of BR and STP in inducing apoptosis, the attention was directed to the activation of cell death processes through various cell cycle signals. This is the first report describing in vivo antitumor activity of BR and its utilization in combination chemotherapy.
Collapse
Affiliation(s)
- P Rauko
- Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, SK-833 91 Bratislava, Slovak Republic
| | | | | | | | | | | |
Collapse
|
9
|
Santell RC, Kieu N, Helferich WG. Genistein inhibits growth of estrogen-independent human breast cancer cells in culture but not in athymic mice. J Nutr 2000; 130:1665-9. [PMID: 10867033 DOI: 10.1093/jn/130.7.1665] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The studies presented were conducted to assess the effect of the soy isoflavone genistein on proliferation of estrogen-independent human breast cancer cells (MDA-MB-231) in vitro and in vivo. Genistein (20 mcmol/L) inhibited cell proliferation in vitro by approximately 50%. Cell cycle progression was blocked in G(2)/M with 40 and 80 mcmol/L genistein. To evaluate the effect of dietary genistein on tumor growth in vivo, genistein was fed to female athymic mice inoculated with MDA-MB-231 cells. After solid tumor masses had formed, mice were fed genistein at a dose (750 mcg/g AIN-93G diet), shown to produce a total plasma genistein concentration of approximately 1 mcmol/L. This dose of genistein did not significantly (P > 0.05) alter tumor growth. Studies were then conducted to assess the effect of dietary genistein on initial tumor development and growth. Genistein (750 mcg/g AIN-93G diet), fed 3 d before cells were inoculated into mice, did not significantly (P > 0.05) inhibit tumor formation or growth. The plasma concentration of genistein in mice fed this dose of dietary genistein (750 mcg/g AIN-93G diet) does not appear sufficient to inhibit tumor formation or growth. Dietary genistein at 750 mcg/g AIN-93G diet does not inhibit tumor formation or growth. Additional studies were conducted to determine the effect of dietary dosages ranging from 0 to 6000 mcg/g AIN-93G diet on plasma genistein concentration. Plasma genistein concentration increased in a dose-dependent manner up to 7 mcmol/L at 6000 mcg/g AIN-93G diet. These data suggest that although genistein inhibits cancer cell growth in vitro, it is unlikely that the plasma concentration required to inhibit cancer cell growth in vivo can be achieved from a dietary dosage of genistein.
Collapse
Affiliation(s)
- R C Santell
- Department of Food Science and Human Nutrition, Michigan State University, E. Lansing, MI 48824, USA
| | | | | |
Collapse
|
10
|
Lian F, Bhuiyan M, Li YW, Wall N, Kraut M, Sarkar FH. Genistein-induced G2-M arrest, p21WAF1 upregulation, and apoptosis in a non-small-cell lung cancer cell line. Nutr Cancer 1998; 31:184-91. [PMID: 9795970 DOI: 10.1080/01635589809514701] [Citation(s) in RCA: 116] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Lung cancer is the leading cause of cancer-related death in the world, with increasing incidence in many developed countries. Epidemiological data suggest that consumption of soy products (the isoflavone genistein) may be associated with a decreased risk of breast and prostate cancer; however, such studies are not available for lung cancer. We investigated cell growth inhibition, modulation in gene expression, and induction of apoptosis by genistein in H460 non-small lung cancer cells. Genistein inhibited H460 cell growth in a dose-dependent manner. Flow-cytometric analysis showed that 30 microM genistein arrested cell cycle progression at the G2-M phase. 4,6-Diamidino-2-phenylindole staining, flow-cytometric analysis, and DNA laddering were used to investigate apoptotic cell death, and the results show that 30 microM genistein can cause typical DNA laddering, a hallmark for apoptosis. In addition, flow cytometry and 4,6-diamidino-2-phenylindole staining showed induction of apoptosis by genistein. Our investigation also demonstrated the modulation of p21WAF1 by Western blot analysis of cell lysates obtained from cultured cells treated with 30 and 50 microM genistein for 24, 48, and 72 hours. Simultaneously, immunocytochemical staining was conducted for the expression of p21WAF1 protein. Our results showed that genistein can upregulate p21WAF1 expression in genistein-treated cells. From these results, we conclude that genistein may act as an anticancer agent, and further studies may prove its efficacy in non-small lung cancer cells. Thus the biological effects of genistein may, indeed, be due to the modulation of cell growth, cell death, and cell cycle regulatory molecules.
Collapse
Affiliation(s)
- F Lian
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | | | |
Collapse
|
11
|
Weber G, Shen F, Li W, Prajda N, Hullan L, Yang H. Novel advances in the regulation of signal transduction activity. ADVANCES IN ENZYME REGULATION 1998; 38:49-62. [PMID: 9762346 DOI: 10.1016/s0065-2571(98)00002-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- G Weber
- Laboratory for Experimental Oncology, Indiana University School of Medicine, Indianapolis 46202-5119, USA
| | | | | | | | | | | |
Collapse
|
12
|
Gozlan J, Lathey JL, Spector SA. Human immunodeficiency virus type 1 induction mediated by genistein is linked to cell cycle arrest in G2. J Virol 1998; 72:8174-80. [PMID: 9733859 PMCID: PMC110162 DOI: 10.1128/jvi.72.10.8174-8180.1998] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Protein tyrosine kinase (PTK) phosphorylation is involved in cellular proliferation and differentiation processes that are key factors for human immunodeficiency virus type 1 (HIV-1) regulation in infected monocytic cells. Short-term exposure of the chronically infected promyelocytic OM10 cell line with the PTK inhibitor genistein induced a dose-dependent increase in p24 antigen production in culture supernatants. This induction persisted in the presence of the reverse transcriptase inhibitor, zidovudine, and was associated with an increased transcription of HIV-1 multiply spliced and unspliced RNAs, suggesting a transcriptional mechanism targeting the integrated provirus. Genistein induced cell differentiation, apoptosis, and a G2 arrest in the OM10 cells. Cell differentiation and apoptosis were not directly involved in the observed increase in HIV-1 replication that was closely linked to genistein-induced G2 arrest. Alleviation of the G2 arrest by pentoxyfylline resulted in a concomitant reduction of HIV-1 to baseline replication. Additionally, by flow cytometry, a significant increase in the number of p24 antigen-expressing cells was observed in cells arrested in G2 compared to those located in G1 or S. Tyrosine kinase inhibition was found not to be essential for enhanced viral replication, which seemed to be related to two other properties of genistein, inhibition of topoisomerase II activity and inhibition of phosphotidylinositol turnover. These findings are consistent with the recent observation that HIV-1 Vpr induces viral replication through preventing proliferation of cells by arresting them in G2 of the cell cycle and strongly suggest that manipulation of the cell cycle plays an important role in HIV-1 pathogenesis.
Collapse
Affiliation(s)
- J Gozlan
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | | | | |
Collapse
|
13
|
Chen HW, Huang HC. Effect of curcumin on cell cycle progression and apoptosis in vascular smooth muscle cells. Br J Pharmacol 1998; 124:1029-40. [PMID: 9720770 PMCID: PMC1565483 DOI: 10.1038/sj.bjp.0701914] [Citation(s) in RCA: 150] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
1. The possible mechanisms of the antiproliferative and apoptotic effects of curcumin (diferuloylmethane), a polyphenol in the spice turmeric, on vascular smooth muscle cells were studied in rat aortic smooth muscle cell line (A7r5). 2. The proliferative response was determined from the uptake of [3H]-thymidine. Curcumin (10(-6)-10(-4) M) inhibited serum-stimulated [3H]-thymidine incorporation of both A7r5 cells and rabbit cultured vascular smooth muscle cells in a concentration-dependent manner. Cell viability, as determined by the trypan blue dye exclusion method, was unaffected by curcumin at the concentration range 10(-6) to 10(-5) M in A7r5 cells. However, the number of viable cells after 10(-4) M curcumin treatment was less than the basal value (2 x 10(5) cells). 3. To analyse the various stages of the cell cycle, [3H]-thymidine incorporation into DNA was determined every 3 h. After stimulation with foetal calf serum, quiescent A7r5 cells started DNA synthesis in 9 to 12 h (G1/S phase), then reached a maximum at 15 to 18 h (S phase). Curcumin (10(-6)-10(-4) M) added during either the G1/S phase or S phase significantly inhibited [3H]-thymidine incorporation. 4. Following curcumin (10(-6)-10(-4) M) treatment, cell cycle analysis utilizing flow cytometry of propidium iodide stained cells revealed a G0/G1 arrest and a reduction in the percentage of cells in S phase. Curcumin at 10(-4) M also induced cell apoptosis. It is suggested that curcumin arrested cell proliferation and induced cell apoptosis, and hence reduced the [3H]-thymidine incorporation. 5. The apoptotic effect of 10(-4) M curcumin was also demonstrated by haematoxylin-eosin staining, TdT-mediated dUTP nick end labelling (TUNEL), and DNA laddering. Curcumin (10(-4) M) induced cell shrinkage, chromatin condensation, and DNA fragmentation. 6. The membranous protein tyrosine kinase activity stimulated by serum in A7r5 cells was significantly reduced by curcumin at the concentration range 10(-5) to 10(-4) M. On the other hand, the cytosolic protein kinase C activity stimulated by phorbol ester was reduced by 10(-4) M curcumin, but unaffected by lower concentrations (10(-6)-10(-5) M). 7. The levels of c-myc, p53 and bcl-2 mRNA were analysed using a reverse transcription-polymerase chain reaction (RT-PCR) technique. The level of c-myc mRNA was significantly reduced by curcumin (10(-5)-10(-4) M) treatment. And, the level of bcl-2 mRNA was significantly reduced by 10(-4) M curcumin. However, the alteration of the p53 mRNA level by curcumin (10(-5)-10(-4) M) treatment did not achieve significance. The effects of curcumin on the levels of c-myc and bcl-2 mRNA were then confirmed by Northern blotting. 8. Our results demonstrate that curcumin inhibited cell proliferation, arrested the cell cycle progression and induced cell apoptosis in vascular smooth muscle cells. Curcumin may be useful as a template for the development of drugs to prevent the pathological changes of atherosclerosis and post-angioplasty restenosis. Our results suggest that the antiproliferative effect of curcumin may partly be mediated through inhibition of protein tyrosine kinase activity and c-myc mRNA expression. And, the apoptotic effect may partly be mediated through inhibition of protein tyrosine kinase activity, protein kinase C activity, c-myc mRNA expression and bcl-2 mRNA expression.
Collapse
Affiliation(s)
- H W Chen
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei
| | | |
Collapse
|
14
|
Abstract
Innovative therapies for chronic myelogenous leukemia (CML) have focused mainly on combining autologous transplantation with another modality of therapy for purging of the graft or treatment of the patient after transplant. Of the three categories of innovative therapies, two are based on studies that demonstrate the bcr/abl gene rearrangement in the pathogenesis of CML, whereas the third is based on the observation that allogeneic disparity is important to maintain remissions in CML. The rationale and data supporting these innovative approaches are reviewed in this article and future strategies are discussed.
Collapse
Affiliation(s)
- J S Miller
- Department of Medicine, University of Minnesota, Minneapolis, USA
| |
Collapse
|
15
|
Barnes S, Peterson TG, Coward L. Rationale for the use of genistein-containing soy matrices in chemoprevention trials for breast and prostate cancer. J Cell Biochem 1995; 22:181-7. [PMID: 8538197 DOI: 10.1002/jcb.240590823] [Citation(s) in RCA: 183] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pharmacologists have realized that tyrosine kinase inhibitors (TKI) have potential as anti-cancer agents, both in prevention and therapy protocols. Nonetheless, concern about the risk of toxicity caused by synthetic TKIs restricted their development as chemoprevention agents. However, a naturally occurring TKI (the isoflavone genistein) in soy was discovered in 1987. The concentration of genistein in most soy food materials ranges from 1-2 mg/g. Oriental populations, who have low rates of breast and prostate cancer, consume 20-80 mg of genistein/day, almost entirely derived from soy, whereas the dietary intake of genistein in the US is only 1-3 mg/day. Chronic use of genistein as a chemopreventive agent has an advantage over synthetic TKIs because it is naturally found in soy foods. It could be delivered either in a purified state as a pill (to high-risk, motivated patient groups), or in the form of soy foods or soy-containing foods. Delivery of genistein in soy foods is more economically viable ($1.50 for a daily dose of 50 mg) than purified material ($5/day) and would require no prior approval by the FDA. Accordingly, investigators at several different sites have begun or are planning chemoprevention trials using a soy beverage product based on SUPRO, an isolated soy protein manufactured by Protein Technologies International of St. Louis, MO. These investigators are examining the effect of the soy beverage on surrogate intermediate endpoint biomarkers (SIEBs) in patients at risk for breast and colon cancer, defining potential SIEBs in patients at risk for prostate cancer, and determining whether the soy beverage reduces the incidence of cancer recurrence.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- S Barnes
- Department of Pharmacology, University of Alabama at Birmingham 35294, USA
| | | | | |
Collapse
|