Gastric Cancer Open Access
Copyright ©The Author(s) 2002. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Apr 15, 2002; 8(2): 224-229
Published online Apr 15, 2002. doi: 10.3748/wjg.v8.i2.224
Effect of cis-9, trans-11-conjugated linoleic acid on cell cycle of gastric adenocarcinoma cell line (SGC-7901)
Jia-Ren Liu, Bai-Xiang Li, Bing-Qing Chen, Yan-Mei Yang, Yu-Mei Zheng, Ying-ben Xue, Department of Toxicological Health, Public Health College, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
Xiao-Hui Han, ICU of Cardiological Surgery, The Second Hospital, Harbin Medical University, Harbin 150001, Heilongjiang Province, China
Rui-Hai Liu, Food Science and Toxicology, Department of Food Science, Cornell University, Ithaca, NY 14853-7201, USA
Author contributions: All authors contributed equally to the work.
Supported by the National Natural Science Foundation of China, No. 39870661
Correspondence to: Dr. Jia-ren Liu, 199 Dongdazhi Street, Nangang District, Harbin 150001, Heilongjiang Province, China.jiarenliu@yahoo.com
Telephone: +86-451-3639459 Fax: +86-451-3641253
Received: August 23, 2001
Revised: September 1, 2001
Accepted: September 5, 2001
Published online: April 15, 2002

Abstract

AIM: To determine the effect of cis-9, trans-11-conjugated linoleic acid (c9,t11-CLA) on the cell cycle of gastric cancer cells (SGC-7901) and its possible mechanism in inhibition cancer growth.

METHODS: Using cell culture and immunocytochemical techniques, we examined the cell growth, DNA synthesis, expression of PCNA, cyclin A, B1, D1, P16ink4a and P21cip/waf1 of SGC-7901 cells which were treated with various c9,t11-CLA concentrations (25, 50, 100 and 200 μmol•L⁻¹)of c9,t11-CLA for 24 and 48 h, with a negative control (0.1% ethane).

RESULTS: The cell growth and DNA synthesis of SGC-7901 cells were inhibited by c9,t11-CLA. SGC-7901 cells. Eight day after treatment with various concentrations of c9,t11-CLA mentioned above, the inhibition rates were 5.92%, 20.15%, 75.61% and 82.44%, respectively and inhibitory effect of c9,t11-CLA on DNA synthesis (except for 25 μmol/L, 24 h) showed significantly less 3H-TdR incorporation than that in the negative controls (P < 0.05 and P < 0.01). Immunocytochemical staining demonstrated that SGC-7901 cells preincubated in media supplemented with different c9,t11-CLA concentrations at various times significantly decreased the expressions of PCNA (the expression rates were 7.2%-3.0%, 24 h and 9.1%-0.9% at 48 h, respectively), Cyclin A (11.0%-2.3%, 24 h and 8.5%-0.5%, 48 h), B1 (4.8%-1.8% at 24 h and 5.5%-0.6% at 48 h)and D1 (3.6%-1.4% at 24 h and 3.7%-0% at 48 h) as compared with those in the negative controls (the expressions of PCNA, Cyclin A, B1 and D1 were 6.5% at 24 h and 9.0% at 48 h, 4.2% at 24 h and 5.1% at 48 h, 9.5% at 24 h and 6.0% at 48 h, respectively) (P < 0.01), whereas the expressions of P16ink4a and P21cip/waf1, cyclin-dependent kinases inhibitors (CDKI), were increased.

CONCLUSION: The cell growth and proliferation of SGC-7901 cell is inhibited by c9,t11-CLA via blocking the cell cycle, with reduced expressions of cyclin A, B1 and D1 and enhanced expressions of CDKI (P16ink4a and P21cip/waf1).




INTRODUCTION

Gastric cancer is common in China[1-12], and it is currently thought to be caused by environmental factors, with diet being an important modifying agent[13-17]. Its mechanism of prevention and treatment still makes it become a hot spot in this area[18-39]. Its anticancerous potential Dietary fat has been implicated as an enhancing agent in carcinogenesis by both epidemiological and animal studies. Consumption of meat, specifically animal fat, has been implicated in a number of disease processes[40-42]. However, several epidemiological studies have suggested an association between increased consumption of meat and fat and decreased risk of stomach, mammary and esophageal cancers[43,44]. Among the fatty acids, only the essential fatty acid, linoleic acid (LA), has been clearly shown to enhance mammary tumorigenesis[43]. However, isomeric derivatives of cis-9, cis-12-octadecadienoic acid (linoleic acid, LA) containing a conjugated double-bond system (conjugated linoleic acid, CLA) showed inhibitory effect on carcinogenesis in animal studies[45,46]. CLA has a mixture of positional (9/11 or 10/12 double bonds) and geometric (various cis/trans combinations) isomers of LA formed by rumen and colon bacteria. The ability of CLA to prevent mammary and other tumors in rodents has been identified and has been the subject of several reviews[43]. There are eight potential isomers of CLA, but the cis-9, trans-11 and trans-9, cis-11 isomers are thought to be active as potential antioxidant and anticarcinogenic agents. Therefore, it is of interest to investigate more extensively the anticancer activities of CLA.

In the present study, we investigated the effect of cis -9, trans -11-CLA (c9,t11-CLA) on the cell cycle of human gastric adenocarcinoma cells (SGC-7901).

MATERIALS AND METHODS
Materials

c9,t11-CLA, a monoisomer of c -9, t 11-octadecadienoic acid with 98% purity, was obtained from Dr. Rui-Hai Liu (Food Science and Toxicology, Department of Food Science, Cornell University, Ithaca, NY, USA). The c9,t11-CLA was dissolved in 96 mL•L⁻¹ ethanol, and was then diluted to the following concentrations: 0, 25, 50, 100 and 200 μmol•L⁻¹.

Methods

Cell culture Human gastric adenocarcinoma cells (SGC-7901), purchased from Cancer Research Institute of Beijing (China), were cultured in RPMI 1640 (Gibco) medium, supplemented with calf serum 100 mL•L⁻¹, penicillin (100 × 103 U•L⁻¹) and streptomycin (100 mg•L⁻¹). The pH was maintained at 7.2-7.4, by equilibration with 5% CO2.The temperature was maintained at 37 °C.The cells were sub-cultured with a mixture Ethylenedinitrile tetraacetic acid (EDTA) and trypsin.

Cell growth curve The SGC-7901 cells were seeded in six 24 well plates (Nuc, Co.); each well contained 2 × 104 cells. After 24 h, the medium of different plates was replaced with media supplemented with c9,t11-CLA at different concentrations. On the next day, the numbers of cells of 3 wells from each plate were determined daily by using the trypan blue staining. The means were obtained on each of eight days and were used to draw a cellular growth curve. The inhibitory rates (IR) on the 8thday was calculated, as follows:

IR (%) = [Total Number of cells in negative control (8 d) - Number of cells in test groups (8 d)]/[Total number of cells in negative control (8 d)] × 100%

[3H]-Labeled precursor incorporation SGC-7901 cells (5 × 104/well in 24 well plate) were cultured in appropriate medium for 24 h prior to beginning the experiment. The medium was, then, replaced with different concentrations c9,t11-CLA. After 18 and 42 h, the cells were incubated with [3H] thymidine (China Nucleus Institute, 0.5 μci/mL, 1.0 μci/well). After 6 h the cells were harvested with trysin/EDTA. Cells were collected in an acetic fiber filter with cellular collector and washed three times with PBS. The filter was dried overnight at 37 °C. The filter was transferred into liquid of scintillation (containing 1% po and 2% pop in xylene) and cpm value determined by liquid Scintillation Counter (LS6500, Beckmen Co.).

Cell samples

SGC-7901 cells were treated for 24 and 48 h with various concentrations of c9,t11-CLA and collected by centrifugation. Specimens were fixed immediately in 40 g•L⁻¹ formaldehydum polymerisatum and embedded in paraffin. Gastric cancer tissue from a patient served as a reference.

Primary antibodyies

To examine the proliferating cell nuclear antigen (PCNA) in cell proliferation and to determine cyclins (A, B1 and D1) and cyclin-dependent kinases inhibitors (P16ink4a and P21waf1) in cell cycle of SGC-7901, we used six primary antibodies: corresponding mouse monoclonal antibodies for cyclin B1 and D1, PCNA and P21waf1 and corresponding rabbit polyclonal antibodies for cyclin A and P16ink4a. PCNA, P16ink4a, and cyclin D1 were purchased from Calbiochem Co.USA; others from Zhongshan Co. China.

Immunocytochemistry

Immunocytochemical staining was performed on serial sections at room temperature, using the horseradish peroxidase method. The sections were deparaffinized in xylene and rehydrated through graded alcohol. The sections were incubated for 10 min at 95 °C in 10 mmol•L⁻¹ sodium citrate (pH6.0) buffer for PCNA staining. Endogenous peroxidases were inactivated by immersing the sections in hydrogen peroxide for 10 min, and then were incubated for 10 min with 100 mL•L⁻¹ normal goat serum in PBS to block non-specific binding. The sections were subsequently incubated overnight at 4 °C with relevant antibodies (1∶50 dilution) respectively. The next day, the sections were incubated with biotinylated anti-mouse or anti-rabbit IgG (Zhongshan Co. China) for 30 min, followed by peroxidase-conjugated streptavidin (Zhongshan Co.China) for 30 min. The chromogenic reaction was developed with DAB (diaminobenzidine) for 10 min, and all sections were counterstained with hematoxylin. Controls consisted of omission of the primary antibody. The Positive Rate (PR) was calculated as follows:

PR (%) = (Number of positive cells)/[Total number (2 × 104)] × 100

Statistical analysis

Analysis of data was performed using the student's t test or χ² test. A value of P < 0.05 is considered to be statistically significant.

RESULTS
Effect of c9,t11-CLA on SGC-7901 cell growth

As shown in Figure 1, Growth of the cells in various concentrations of c9,t11-CLA did not differ from the negative control within 3 d. After 3 d, SGC-7901 cells incubated in 25 and 50 μmol•L⁻¹ of c9,t11-CLA grew at a lower rate than the negative control. While in 100 and 200 μmol•L⁻¹ concentrations of c9,t11-CLA, cell proliferation was significantly inhibited. The inhibitory rate of various c9,t11-CLA concentrations were 5.9%, 20.2%, 75.6% and 82.4%, respectively.

Figure 1
Figure 1 Growth curve of SGC-7901 cells cultured in various concentration of c9,t11-CLA
Effect on DNA synthesis

The effect of CLA on isotope incorporation into SGC-7901 cells are presented in Table 1. SGC-7901 cells preincubated in media supplemented with various c9,t11-CLA concentration (except for 25 μmol/L, 24 h) incorporated significantly less 3H-TdR than did the negative control (P < 0.05 and P < 0.01, Table 1). The inhibitory rate (IR) displayed a dose-response relationship as the concentration of c9,t11-CLA increased.

Table 1 Inhibitory effect of c9,t11-CLA on DNA synthesis in SGC-7901 cells (n = 6).
c9,t11-CLA (μmol/L)3H-TdR incorporation (cpm, ¯x ± s)
Inhibitory Rate (%)
24 h48 h24 h48 h
02165 ± 1723598 ± 603
251810 ± 5053093 ± 32316.414.0
502208 ± 2912640 ± 607a-2.026.6
1002065 ± 2612063 ± 495b4.642.7
200472 ± 260b88 ± 15b78.297.5
Cell proliferation

As shown in Figure 2, expression raues of PCNA (Figure 3) on SGC-7901 cells gradually decreased after SGC-7901 cells were incubated with different concentrations of c9,t11-CLA at various times. Moreover, SGC-7901s cell expressed significantly less PCNA than did the negative control (P < 0.01). The expression rate displayed a dose-response relationship as the concentrations of CLA increased.

Figure 2
Figure 2 Expression of PCNA on SGC-7901 cells treated with c9,t11-CLA
Figure 3
Figure 3 A: The expression of PCNA on SGC-7901 cells of the negative controls (immunocytochemistry staining SP method, original magnification × 400); B: The expression of cyclin A on SGC-7901 cells of the negative controls (immunocytochemistry staining SP method, original magnification × 400); C: The expression of cyclin B1 on SGC-7901 cells of the negative controls (immunocytochemistry staining SP method, original magnification × 400); D: The expression of cyclin D1 on SGC-7901 cells of the negative controls (immunocytochemistry staining SP method, original magnification × 400); E: The expression of P16inf4aon SGC-7901 cells of c9,t11-CLA group (100 μmol•L⁻¹) (immunocytochemistry staining SP method, original magnification × 400); F: The expression of P21cip/waf1 on SGC-7901 cells of c9,t11-CLA group (100 μmol•L⁻¹) (immunocytochemistry staining SP method, original magnification × 400)
Expressions of cyclin A, B1 and D1 and P16ink4a, p21waf1

The the expression of rates cyclin A, B1, and D1 (Figure 3)on SGC-7901 cells was decreased (Table 2) after SGC-7901 cell were incubated with different concentrations of c9,t11-CLA for 24 h and 48 h while cyclin-dependent kinases inhibitors (P16ink4a, and P21waf1) increased (Table 3; Figure 3).

Table 2 Positive rates of cyclin A, B1, and D1 on SGC-7901 cells treated with c9,t11-CLA (%).
c9,t11-CLA (μmol/L)24 h
48 h
Cylin ACylin B1Cyclin D1Cylin ACylin B1Cyclin D1
010.74.29.55.95.16.0
2511.04.83.6b8.55.53.7b
507.92.53.5b5.03.1b3.7b
1004.4b2.6b2.1b1.3b0.7b0.6b
2002.3b1.8b0.4b0.5b0.6b0
Table 3 Positive rates of p16ink4a and p21waf1 on SGC-7901 cells treated with c9,t11-CLA (%).
c9,t11-CLA (μmol/L)24 h
48 h
p16ink4ap21waf1p16ink4ap21waf1
01.00.20.80.6
250.71.4b0.20.8
501.41.0b3.0b2.5b
1002.8b4.1b4.6b3.8b
2003.6b5.2b5.0b6.3b
DISCUSSION

CLA is a naturally occurring fatty acid in animal's food. Dietary sources of CLA include grilled beef, cheese, and related foods[47]. Another source of CLA is its endogenous generation via the carbon centered free radical oxidation of linoleic acid[45]. Over the past ten years, a number of research works of animal experiments have supported the observation that CLA is an effective chemopreventive agent of cancer, and that it can inhibit carcinogenesis of different tissues at different stages of induction by chemical agents[44,45]. Several investigators in our group have reported that c9,t11-CLA is an effective agent to prevent carcinogenesis[48,49] and cancer[50-52]. Zhu's study[48] demonstrated that c9,t11-CLA could significantly inhibit the mice forestomach neoplasia induced by B(a)P (50 mg·kg-1) in post-initiation in short term (23weeks).The incidences of tumors of mice in the B(a)P group, B(a)P with high dose CLA (5 μL·g-1) group and B(a)P with low dose CLA (2.5 μL·g-1) group were 100%, 60% and 69% respectively (P < 0.05). Xue's research[49] also indicated that the incidence of neoplasm in mouse forestomach in the B(a)P group, 75% pure c9,t11-CLA group, 98% pure c9,t11-CLA group and 98% pure t10, c12-CLA group were 100.0%, 75.0%, 69.2%, and 53.8%, respectively. This maybe due to an inhibition mitogen of activated protein kinase (MAPK)-a way to reduce carcinogenesis. The data from our research group suggested that c9,t11-CLA could inhibit proliferation of cancer cells, i.e. SGC-7901 cells[50] and MCF-7 cells[51,52], and induced cancer cell (SGC-7901) apoptosis[53]. Moreover, the inhibiting effect of c9,t11-CLA on SGC-7901 cell proliferation may be related to cell cycle.

As shown in Figure 1, c9,t11-CLA at various concentrations in 8 d reduced the proliferative activity of SGC-7901 cells and its inhibitory rates were from 5.92% to 82.44%, but the mechanism of such inhibition of c9,t11-CLA has not been clarified. However, we discovered that SGC-7901 cells supplemented with c9,t11-CLA incorporated significantly less [3H] thymidine than negative controls (shown in Table I). The inhibitory rates of, from 16.4% to 78.2% after incubating with c9,t11-CLA for 24 h and from 14.0% to 97.5% after 48 h displayed a dose-response relationship.In the meantime, we investigated further the expressions of PCNA and protein from cell cycle such as cyclins and cyclin-dependent kinase inhibitors (CDKI) on SGC-7901 cells treated with various concentrations of c9,t11-CLA. PCNA (proliferating cell nuclear antigen) plays an essential role in both the replication and repair of DNA, and is an essential component of the DNA replication machinery, acting as the processing factor for polymerases δ and ε. In addition to its role in replication, PCNA is not only required for base excision-repair of nucleotides, but also binds to cell cycle regulatory proteins such as P21 and Gadd45[54]. In this study, we discovered that the expression of PCNA on SGC-7901 cells gradually decreased with increasing concentrations of c9,t11-CLA in comparison with negative controls (showed in Figure 2). In other words, DNA replication lessens, thereby resulting in slower on SGC-7901 cell proliferation.

The fundamental task of the cell cycle is to ensure that DNA is faithfully replicated once during S phase and that identical chromosomal copies are distributed equally to two daughter cells during M phase. The cell cycle is a complex process, regulated by many factors, which can be divided into three groups: cyclins (A, B, D, E....H); cyclin-dependent kinases (CDK, including CDK1-CDKa-7); CDK inhibitors (CDKI, including P16 family and P21 family). They are balanced through mutual interactions. Uncontrolled cell proliferation is the hallmark of cancers which are the result of damage to genes that directly regulate their cell cycles. Using immunocytochemical technique to detect expressions of cyclins and CDKI, we demonstrated that the expressions of cyclin A, B1 and D1 on SGC-7901 cells treated with various concentrations c9,t11-CLA were reduced, whereas expressions of CDKI (P16in4a and P21waf1) increased, as compared with those of negative controls. Successive actions of CDKs promote cell-cycle progression in mammalian cells. Various cyclins bind and activate CDKs at specific times during the cell cycle.

Mammalian cyclin A activates CDK2[55] in S-phase and CDK1 (Cdc2) in G2- and M-phases. One important mechanism that enables sequential activation of cyclin-CDK complexes is the periodic synthesis and destruction of cyclins. Cyclin A expression starts late in G1-phase and is increasing through S- and G2-phase before the protein is degraded in M-phase. The cell cycle-dependent expression of cyclin B1 is critical for the proper timing of a cell's entry into mitosis which is dependent both upon the binding of CDK1 to cyclin B1, as well as a series of phosphorylation and dephosphorylation events. The cyclin B1 protein accumulates during interphase and peaks at the G2-M phase transition[56]. One of the crucial substrates of G1 phase CDK, including CDK4 in the complex with D-type cyclins (cyclin D1, D2 and D3), is Rb protein (pRb), which is the product of the retinoblastoma susceptibility gene. Rb protein plays an important role in the regulation of the G1 to S phase progression in normal cells and the function of pRb is regulated by phosphorylation. Thus, during the G0 and G1 phase, Rb protein is in an un- or underphosphorylated stage and binds to E2F family transcription factors. Cylin Ds/CDK4 becomes activated around the mid G1 phase, resulting in the accumulation of increasingly phosphorylated, inactive forms pRb. This causes the release of E2F family transcription factors which induce the expression of S-phase genes by positive regulation through E2F-binding sites (see Figure 4)[57]. It is also known that abrogation of the functions of Cylin A prevents entry into the S phase. From the beginning of the S phase Rb protein remains in the hyperphosphorylated inactive state until the end of M phase; such condition is thought to be due to both cylin A/CDK2 and Cylin A, B1/Cdc2 in catalyzing the phosphorylation reaction[58]. P16ink4a is the founder member of a family of proteins with the ability to inhibit CDK4 and the CDK4-related kinase CDK6.

Figure 4
Figure 4 The relationship between CDKI (P16 and P21) and cyclins in G1/S transition

The INK4 family is composed of four members in mammalian organisms: P16ink4a, P15ink4b, P18ink4c, and P19ink4 d. The four mammalian INK4 proteins have similar biochemical properties: All of them bind to CDK4 and CDK6 and inhibit the kinase activity of the CDK4-6/Cylin D complexes (Figure 4)[59]. The INK4 inhibitor causes G1 arrest indicating that the phosphorylation of pRb on residues specific for CDK4 (and possibly CDK6) is critical for G1/S progression. While P21CIP1/waf1family, comprisingP21Cip1/waf1, P27CIP1 and P57CIP2, bind to a variety of CDKs and cyclins, preferentially to cyclin/CDK complexes rather than monomeric forms and also inhibit performed active cyclin/CDK complexes (see Figure 4)[59]. In addition to its role as a CDKI, P21Cip/waf1 has been shown to block DNA replication by direct interaction with PCNA mentioned above. However, P21CIP1/waf1 does not inhibit the PCNA-dependent nucleotide excision-repair of DNA. In deed, DNA damage leads to an increase in the level of P53, and result in P21-mediated cell cycle arrest in the G1 phase, which persists until DNA repair is completed[60].Thus, it is proposed that P21Cip/waf1 plays an important role under such conditions as terminal differentiation and cell senescence.

In conclusion, c9,t11-CLA may inhibit cell growth and proliferation by a decrease in the expressions of cyclin A, B1 and D1 and an increase in that of CDKI (P16ink4a and P21Cip1/waf1) on SGC-7901 cells in comparison with the negative controls. This result suggested that the inhibition effect of c9,t11-CLA on SGC-7901 cell proliferation is related to the cell cycle. The whole mechanism of the action of c9,t11-CLA on SGC-7901 cell cycle further research.

Footnotes

Edited by Lu HM

References
1.  Gao HJ, Yu LZ, Bai JF, Peng YS, Sun G, Zhao HL, Miu K, L XZ, Zhang XY, Zhao ZQ. Multiple genetic alterations and behavior of cellular biology in gastric cancer and other gastric mucosal lesions: H. pylori infection, histological types and staging. World J Gastroenterol. 2000;6:848-854.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Deng DJ, E Z. Overview on recent studies of gastric carcinogenesis: human exposure of N nitrosamides. Shijie Huaren Xiaohua Zazhi. 2000;8:250-252.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Deng DJ. progress of gastric cancer etiology: N-nitrosamides 1999s. World J Gastroenterol. 2000;6:613-618.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Li DG, Wang ZR, Lu HM. Pharmacology of tetrandrine and its therapeutic use in digestive diseases. World J Gastroenterol. 2001;7:627-629.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Niu WX, Qin XY, Liu H, Wang CP. Clinicopathological analysis of patients with gastric cancer in 1200 cases. World J Gastroenterol. 2001;7:281-284.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Kimura K. Gastritis and gastric cancer. Asia. Gastroenterol Clin North Am. 2000;29:609-621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
7.  Hua JS. Effect of Hp: cell proliferation and apoptosis on stomach cancer. Shijie Huaren Xiaohua Zazhi. 1999;7:647-648.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Xia HX, Zhang GS. Apoptosis and proliferation in gastric cancer caused by Hp infection. Shijie Huaren Xiaohua Zazhi. 1999;7:740-742.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Cai L, Yu SZ, Ye WM, Yi YN. Fish sauce and gastric cancer: An ecological study in Fujian Province, China. World J Gastroenterol. 2000;6:671-675.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Cai L, Yu SZ, Zhang ZF. Helicobacter pylori infection and risk of gastric cancer in Changle County, Fujian Province, China. World J Gastroenterol. 2000;6:374-376.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Cai L, Yu SZ. A molecular epidemiologic study on gastric cancer in Changle, Fujian Province. Shijie Huaren Xiaohua Zazhi. 1999;7:652-655.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Cai L, Yu SZ, Zhang ZF. Glutathione S-transferases M1, T1 genotypes and the risk of gastric cancer: A case-control study. World J Gastroenterol. 2001;7:506-509.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Sugie S, Okamoto K, Watanabe T, Tanaka T, Mori H. Suppressive effect of irsogladine maleate on N-methyl-N-nitro-N-nitrosoguanidine (MNNG)-initiated and glyoxal-promoted gastric carcinogenesis in rats. Toxicology. 2001;166:53-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
14.  Palli D, Russo A, Saieva C, Salvini S, Amorosi A, Decarli A. Dietary and familial determinants of 10-year survival among patients with gastric carcinoma. Cancer. 2000;89:1205-1213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
15.  Mathew A, Gangadharan P, Varghese C, Nair MK. Diet and stomach cancer: A case-control study in South India. Eur J Cancer Prev. 2000;9:89-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 60]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
16.  Palli D. Epidemiology of gastric cancer: An evaluation of available evidence. J Gastroenterol. 2000;35 Suppl 12:84-89.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Palli D, Russo A, Ottini L, Masala G, Saieva C, Amorosi A, Cama A, D'Amico C, Falchetti M, Palmirotta R. Red meat, family history, and increased risk of gastric cancer with microsatellite instability. Cancer Res. 2001;61:5415-5419.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Tu SP, Jiang SH, Tan JH, Jiang XH, Qiao MM, Zhang YP, Wu YL, Wu YX. Proliferation inhibition and apoptosis inductionby arsenic trioxide on gastric cancer cell SGC-7901. Shijie Huaren Xiaohua Zazhi. 1999;7:18-21.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Chen YQ, Shen HX, Zhou SJ, Chen L, Yu X, Lu LP. Experimental study of the inhibitory effect of fufang shenqitang ongastric adenocarcinoma of rats. Shijie Huaren Xiaohua Zazhi. 1999;7:117-119.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Gao GL, Yang Y, Ren CW, Yang SF. Effect of vitamin D3 on forestomach carcinogenesis induced by N methyl N'nitroN nitroso-guanidine and tween 20 in rats. Shijie Huaren Xiaohua Zazhi. 1999;7:763-765.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Xiao B, Xiao LC, Lai ZS, Zhang YL, Zhang ZS, Zhang WD. Experimental study of the inhibition affect of Zhen’ailing onthe growth of gastric cancer in vitro. Shijie Huaren Xiaohua Zazhi. 1999;7:951-954.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Xin Y, Zhao FK, Zhang SM, Wu DY, Wang YP, Xu L. Relationship between CD44v6 expression and prognosis in gastriccarcinoma patients. Shijie Huaren Xiaohua Zazhi. 1999;7:210-214.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Li XS, Zhang XJ, Sun YR, Wang XF, Liu HW, Li W, Sa HJ, Liu HY, Zhang XM, Yang JC. Studies on cytokine activity insera and ascites of patients with digestive tumor. Shijie Huaren Xiaohua Zazhi. 1999;7:13-14.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Wang SW, Xie YH, Li YR, Lin HS, Zhu LZ. Antineoplastic effect and short-term effect on advanced malignant tumor of digestive tract of Chinese herbs antike. Shijie Huaren Xiaohua Zazhi. 1999;7:236-239.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Liu HF, Liu WW, Fang DC, Men RF, Wang ZH. Apoptosis and its relationship with Fas ligand expression in gastric carcinoma and its precancerous lesion. Shijie Huaren Xiaohua Zazhi. 1999;7:561-563.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Zhou HP, Wang X, Zhang NZ. Early apoptosis in intestinal and diffuse gastric carcinomas. World J Gastroenterol. 2000;6:898-901.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Liu XB, Li L, Zhuang BZ, Jiang YD, Wang JH. Cyclin E expression in gastric carcinoma and its clinicopathological significance. Shijie Huaren Xiaohua Zazhi. 1999;7:656-658.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Wang XS, Wang RB, Zhang ZL, Cao TJ, Jin WD. Effect of short time heating with MMC and 5-FU on cancer cells. Shijie Huaren Xiaohua Zazhi. 1999;7:576-578.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Zhang FX, Zhang XY, Fan DM, Deng ZY, Yan Y, Wu HP, Fan JJ. Antisense telomerase RNA induced human gastric cancer cell apoptosis. World J Gastroenterol. 2000;6:430-432.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Zhao AG, Yang JK, Zhao HL. Chinese Jianpi herbs induce apoptosis of human gastric cancer grafted onto nude mice. Shijie Huaren Xiaohua Zazhi. 2000;8:737-740.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Zhu ZH, Xia ZS, He SG. The effects of ATRA and 5Fu on telomerase activity and cell growth of gastric cancer cellsin vitro. Shijie Huaren Xiaohua Zazhi. 2000;8:669-673.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Xia ZS, Zhu ZH, He SG. Effects of ATRA and 5-FU on growth and telomerase activity of xenografts of gastric cancer in nude mice. Shijie Huaren Xiaohua Zazhi. 2000;8:674-677.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Guo YQ, Zhu ZH, Li JF. Flow cytometric analysis of apoptosis and proliferation in gastric cancer and precancerous lesion. Shijie Huaren Xiaohua Zazhi. 2000;8:983-987.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Gu QL, Li NL, Zhu ZG, Yin HR, Lin YZ. A study on arsenic trioxide inducing in vitro apoptosis of gastric cancer cell lines. World J Gastroenterol. 2000;6:435-437.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Wu K, Shan YJ, Zhao Y, Yu JW, Liu BH. Inhibitory effects of RRR-alpha-tocopheryl succinate on benzo (a)pyrene (B(a)P)-induced forestomach carcinogenesis in female mice. World J Gastroenterol. 2001;7:60-65.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Wu K, Liu BH, Zhao DY, Zhao Y. Effect of vitamin E succinate on expression of TGF-beta1, c-Jun and JNK1 in human gastric cancer SGC-7901 cells. World J Gastroenterol. 2001;7:83-87.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Cui RT, Cai G, Yin ZB, Cheng Y, Yang QH, Tian T. Transretinoic acid inhibits rats gastric epithelial dysplasia induced by N-methyl-N-nitro-N-nitrosoguanidine: influences on cell apoptosis and expression of its regulatory genes. World J Gastroenterol. 2001;7:394-398.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  He XS, Su Q, Chen ZC, He XT, Long ZF, Ling H, Zhang LR. Expression, deletion [was deleton] and mutation of p16 gene in human gastric cancer. World J Gastroenterol. 2001;7:515-521.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Liu S, Wu Q, Chen ZM, Su WJ. The effect pathway of retinoic acid through regulation of retinoic acid receptor alpha in gastric cancer cells. World J Gastroenterol. 2001;7:662-666.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Zhang B, Zhang LH, Li N, Wang YJ, Li JS. Effects of n-3 fatty acids on glucose metabolism of septic rats and itsmechanism. Shijie Huaren Xiaohua Zazhi. 1999;7:335-337.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Lewis CJ, Yetley EA. Health claims and observational human data: relation between dietary fat and cancer. Am J Clin Nutr. 1999;69:1357S-1364S.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Mantzioris E, Cleland LG, Gibson RA, Neumann MA, Demasi M, James MJ. Biochemical effects of a diet containing foods enriched with n-3 fatty acids. Am J Clin Nutr. 2000;72:42-48.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  MacDonald HB. Conjugated linoleic acid and disease prevention: A review of current knowledge. J Am Coll Nutr. 2000;19:111S-118S.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 139]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
44.  Kimoto N, Hirose M, Futakuchi M, Iwata T, Kasai M, Shirai T. Site-dependent modulating effects of conjugated fatty acids from safflower oil in a rat two-stage carcinogenesis model in female Sprague-Dawley rats. Cancer Lett. 2001;168:15-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
45.  Ip C, Banni S, Angioni E, Carta G, McGinley J, Thompson HJ, Barbano D, Bauman D. Conjugated linoleic acid-enriched butter fat alters mammary gland morphogenesis and reduces cancer risk in rats. J Nutr. 1999;129:2135-2142.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Brodie AE, Manning VA, Ferguson KR, Jewell DE, Hu CY. Conjugated linoleic acid inhibits differentiation of pre- and post- confluent 3T3-L1 preadipocytes but inhibits cell proliferation only in preconfluent cells. J Nutr. 1999;129:602-606.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Parodi PW. Conjugated linoleic acid and other anticarcinogenic agents of bovine milk fat. J Dairy Sci. 1999;82:1339-1349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 199]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
48.  Zhu Y, Qiu J, Chen BQ, Liu RH. The inhibitory effect of conjugated linoleic acid on mice forestomach neoplasia inducedby benzo (a)pyrene. Zhonghua Yufang Yixue. 2001;35:19-22.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Xue YB, Chen BQ, Liu JR, Zheng YM, Liu RH. The inhibition of mouse forestomach neoplasm induced by B (a)p throughMAPKs pathway by conjugated Linoleic acid. Zhonghua Yufang Yixue. 2001;35:1-4.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Liu J, Chen B, Liu R, Lu G. [Inhibitory effect of conjugated linoleic acid on human gastric carcinoma cell line]. Wei Sheng Yan Jiu. 1999;28:353-355.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Liu JR, Chen BQ, Xue YB, Han XH, Yang YM, Liu RH. Inhibitory effect of conjugated linoleic acid on the in vitro growth ofhuman mammary cancer cells (MCF-7). Zhonghua Yufang Yixue. 2001;35:244-247.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  O'Shea M, Devery R, Lawless F, Murphy J, Stanton C. Milk fat conjugated linoleic acid (CLA) inhibits growth of human mammary MCF-7 cancer cells. Anticancer Res. 2000;20:3591-3601.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Liu JR, Chen BQ, Deng H, Han XH, Liu RH. Cellular apoptosis induced by conjugated linoleic acid in human gastriccancer (SGC-7901) cells. Gongye Weisheng Yu Zhiyebing. 2001;27:129-133.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Tsurimoto T. PCNA binding proteins. Front Biosci. 1999;4:D849-D858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 95]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
55.  Mani S, Wang C, Wu K, Francis R, Pestell R. Cyclin-dependent kinase inhibitors: novel anticancer agents. Expert Opin Investig Drugs. 2000;9:1849-1870.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 66]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
56.  Ito M. Factors controlling cyclin B expression. Plant Mol Biol. 2000;43:677-690.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 59]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
57.  Pyronnet S, Sonenberg N. Cell-cycle-dependent translational control. Curr Opin Genet Dev. 2001;11:13-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 147]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
58.  Viallard JF, Lacombe F, Belloc F, Pellegrin JL, Reiffers J. [Molecular mechanisms controlling the cell cycle: fundamental aspects and implications for oncology]. Cancer Radiother. 2001;5:109-129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 64]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
59.  Israels ED, Israels LG. The cell cycle. Stem Cells. 2001;19:88-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 41]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
60.  Sowa Y, Sakai T. Butyrate as a model for "gene-regulating chemoprevention and chemotherapy.". Biofactors. 2000;12:283-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 44]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]