Colorectal Cancer Open Access
Copyright ©The Author(s) 2003. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Sep 15, 2003; 9(9): 1972-1975
Published online Sep 15, 2003. doi: 10.3748/wjg.v9.i9.1972
Mad2 and p53 expression profiles in colorectal cancer and its clinical significance
Gang-Qiang Li, Hao Li, Hong-Fu Zhang, Department of Pathology, Anhui Medical University, Hefei, 230032, Anhui Province, China
Author contributions: All authors contributed equally to the work.
Supported by Natural Science Fund of Anhui Province, No. 01043717
Correspondence to: Gang-Qiang Li, Department of Pathology, Chinese PLA 455 Hospital, Shanghai 200052, China. lgqfm@163.com
Telephone: +86-21-62800157-2163
Received: March 28, 2003
Revised: April 23, 2003
Accepted: May 21, 2003
Published online: September 15, 2003

Abstract

AIM: To investigate the expression of tumor suppressor gene p53 and spindle checkpoint gene Mad2, and to demonstrate their expression difference in colorectal cancer and normal mucosa and to evaluate its clinical significance.

METHODS: Western blot and immunohistochemistry methods were used to analyze the expression of Mad2 in colorectal cancer and its corresponding normal mucosa. The expression of p53 was detected by immunohistochemistry method in colorectal cancer and its corresponding normal mucosa.

RESULTS: Mad2 was significantly overexpressed in colorectal cancer compared with corresponding normal mucosa (P < 0.001), and it was not related to the differentiation of adenocarcinoma and other clinical factors (P > 0.05).The ratio of Mad2 protein in cancer tissue (C) to that in its normal mucosa tissue (N) was higher than 2, which was more frequently observed in patients with lymph gland metastasis (P < 0.05). p53 protein expression was not observed in normal mucosa. The rate of p53 positive expression in adenocarcinomas was 52.6%. There was a significant difference between adenocarcinomas and normal mucosa(P < 0.001), which was not related to the differentiation degree of adenocarcinoma and other clinical factors (P > 0.05).

CONCLUSION: Defect of spindle checkpoint gene Mad2 and mutation of p53 gene are involved mainly in colorectal carcinogenesis and C/N > 2 is associated with prognosis of colorectal cancer.




INTRODUCTION

Accurate chromosomal segregation is essential for cell survival and genomic stability. Genomic instability is a common feature of human cancer and is thought to be an important contributor to the malignant phenotype[1,2]. At least two categories of instability have been described: CIN and MIN. CIN is characterized by an alteration in chromosome number and is commonly detected as aneuploidy. The fact that the majority of human cancer cells exhibit gains or losses of chromosomes suggests that CIN may contribute to tumorigenesis[3-5]. In yeast, a loss of mitotic checkpoint frequently leads to abnormal chromosome number, resulting in aneuploidy or polyploidy[6]. The mitotic checkpoint, known as the spindle assembly checkpoint, detects errors occurred in the spindle structure or in alignment of the chromosomes on the spindle, and delays chromosome segregation and anaphase onset until the defects are corrected. Two major groups of mitotic checkpoint genes, budding uninhibited by benomyl (BUB) 1-3 and mitotic arrest defect (MAD) 1-3, have been identified in budding yeast[7]. Mammalian homologues of the yeast mitotic checkpoint protein have also been characterized[8-10].

To date, the vast majority of studies about mitotic checkpoint have focused on cancer cell lines. Little data are available on Mad2 in colorectal adenocarcinoma. In this study, the authors used Western blot and immunohistochemical technique to examine the expression of Mad2 and p53 in colorectal cancer to elucidate the relation of Mad2 and p53 to carcinogenesis and clinical pathological factors.

MATERIALS AND METHODS
Specimens

Cancer tissues and corresponding normal tissues were obtained from the First Affiliated Hospital of Anhui Medical University from October 2000 to May 2001. No patient had been treated with anti-neoplasm therapy before tumor removal. Thirty eight patients (21 males, 17 females, aged between 28 to 81 years, median age 54.8 years) were as follows: 25 cases of well differentiated adenocarcinoma, 5 cases of moderately differentiated adenocarcinoma, 8 cases of poorly differentiated adenocarcinoma. Parts of the tissues were embedded in OCT, immediately frozen in liquid nitrogen. The other parts were fixed in 10% formalin, embedded in paraffin, cut into 4 μm serial sections.

Reagents

Mad2 polyclonal rabbit antibodes against human were provided by China Science and Technology University. HEPES, SDS and Polycrylamide were purchased from Sigma Biological Technology Ltd., USA. Immuno-blot nitrocellulose filter membrane was purchased from OSMONICS Biological Technology Ltd., USA. p53, goat anti-rabbit IgG-HRP and S-P immunohistochemical kit were purchased from Beijing Zhongshan Biological Technology Ltd.

Western blot

Both cancer and its corresponding normal frozen tissues were minced and homogenized in buffer (10 mM HEPES, 113 mM mannitol, 37 mM sucrose, 1 mM EDTA) for 5 min on ice. After spun at 15000 rpm for 20 min at 4 °C, supernatants were collected and frozen at -20 °C until use. Protein concentration was measured by modified Lowry protein assay. Samples containing 40 μg total protein were electrophoretically separated and transferred to an immuno-blot nitrocellulose filter membrane. The membranes were blocked overnight at 4 °C with 5% skim milk. The blots were incubated with rabbit polyclonal anti-Mad2 antibody at 1:200 dilution in 5% skim milk for 1 h at 37 °C and were incubated with goat anti-rabbit IgG-HRP at 1:50 dilution in 5% skim milk for 30 min at 37 °C. Following treatment with DAB, the blots were exposed to a gel image system and measured.

Immunohistochemistry

Immunohistochemical staining was performed using S-P method, anti-Mad2 antibody was diluted to 1:120. Anti-p53 was ready to use reagent. A negative control was dyed according to the above method with the primary antibody substituted by animal serum. The standard of positive Mad2 protein expression was stained with brown-yellow color mainly in cell plasma, p53 positive expression was stained in nucleus. A semi-quantitative evaluation was used to determine positively expressed cells by viewing 10 vision fields at × 400[11]. Negative (-) indicated cells were stained less than 10%, mild positive (+) showed 11%-25% cells were stained, moderately positive (++) demonstrated 26%-50% cells was stained, strong positive (+++) revealed over 50% cells were stained. The later three grades were all regarded as positive.

Statistical analysis

The data were analyzed by SPSS version 10.0. Fisher’s exact test or χ2 test was used for statistical analysis. P < 0.05 was considered statistically significant.

RESULTS
Expression of Mad2 protein

We analyzed the results measured by gel image system. Mad2 protein in cancer tissue was significantly overexpressed compared with that in corresponding normal tissues (P < 0.001). (Figure 1)The calculated ratio of Mad2 protein in cancer tissues (C) to that in corresponding normal tissues (N) was 2.07 ± 1.87, ranging from 0.31 to 4.68, indicating that Mad2 expression was approximately 2-fold higher in cancer tissue than in normal tissue. Furthermore, we analyzed the association between the C/N ratio of Mad2 expression and clinicopathological parameters including histological differentiation and lymph node metastasis. The results are summarized in Table 1. No significant difference was found among well differentiated, moderately differentiated, and poorly differentiated adenocarcinomas. However, C/N > 2 was found to be related with lymph node metastasis (P < 0.05).

Table 1 Relationship between C/N ratio of Mad2 expression and histologic differentiation and lymph node metastasis.
GroupsnMad2
Positive (%)P
C/N > 2C/N < 2
Differentiation
WD25101540%
MD52340%
PD82625%aP = 0.737
Lymph node metastasis
Absent3292328%
Present65183.3%bP = 0.01
Figure 1
Figure 1 Mad2 protein expression in normal tissue and colorectal Cancer. 1: standard molecular weight; 2: Mad2 in poorly differentiated adenocarcinoma; 3: Mad2 in correspond-ing norma l tissue; 4: Mad2 in well differ entia ted adenocarcinoma; 5: Mad2 in corresponding normal tissue; 6: Mad2 in moderately differentiated adenocarcinoma; 7: Mad2 in corresponding normal tissue.

The positive signals of Mad2 protein were brown-yellow stains mainly in cell plasma, and strength of color was directly proportional to positive percentage (Figure 2, Figure 3). Positive expression of Mad2 protein was detected in 33 out of 38 (86.8%) colorectal cancers, and 18 out of 38 (47.4%) normal tissues. There was a significant difference between colorectal cancer and normal tissues (P < 0.001). However, no significant difference was found among well differentiated, moderately differentiated, and poorly differentiated adenocarcinomas. The expression of Mad2 in colorectal cancer was not related with lymph node metastasis (Table 2).

Table 2 Relationship between expression of Mad2 protein and histologic differentiation and lymph node metastasis.
GroupsnMad2
Positive (%)P
+-
Normal tissue38182047.4%
Adenocarcinoma3833586.8%aP < 0.001
WD2523292%
MD550100%
PD85362.5%bP = 0.064
Lymph node metastasis
Absent3229390.6%
Present64266.7%cP = 0.169
Figure 2
Figure 2 Strongly positive expression of Mad2 in tubular adenocarcinoma. S-P × 400.
Figure 3
Figure 3 Weakly positive expression of Mad2 in normal mucosa S-P × 400.
Correlation between Mad2 protein and p53

The percentage of positive p53 expression was 52.6% (20/38) in colorectal cancer(Figure 4), no such protein expression was observed in normal tissue. We analyzed the correlation between Mad2 protein and p53 expression by χ2 test. There was a significantly positive correlation between the expressions of Mad2 and p53(P < 0.05).

Figure 4
Figure 4 Strongly positive expression of p53 in tubular adenocarcinoma. S-P × 400.
DISCUSSION

Kinetochores are linked to both chromosomes and microtubules, and play an important role in the generation of mitotic checkpoint signals. The function of kinetochores was to ensure that chromosomes were not segregated until each of them was aligned and attached to the spindle[12,13]. The majority of protein associated with mitotic checkpoint function have been shown to localize at kinetochores unattached to microtubules[14,15]. It has been proposed that the mitotic checkpoint proteins, especially Mad2 be crucial for generating the “wait” signal to prevent the onset of anaphase after microtubule disruption[16-18]. Previous studies indicated that mutations of mitotic checkpoint protein gene or lower expression of mitotic checkpoint protein might lead to the failure of mitotic checkpoint function, which might in turn lead to tumorigenesis, especially aneuploid cancer. Li and benezra reported that one breast cancer cell line with a mitotic checkpoint defect expressed Mad2 at a level one-third less than that of diploid normal cell lines[8]. Recently, Takahashi et al[19] reported mitotic checkpoint function was impaired in 4 (44%) out of 9 human lung cancer cell lines. In the present study, the expressions of Mad2 and p53 protein were examined in colorectal cancer and its corresponding normal tissues. Mad2 expression in colorectal cancer was higher than that in its corresponding normal tissues. Our results on the expression of Mad2 were not consistent with the reduced Mad2 expression in a breast cancer cell[8]. We speculate it might be the surrounding in which cell lived that led to the expression difference. In addition, positive expression of Mad2 was found in normal tissues to some extent. In budding yeast, the amount of Mad2 protein appeared to be constant during the cell cycle, although the level of Clb2p, a mitotic cycle cyclin, changed regularly in response to cell progression[20]. Similarly, a certain amount of Mad2 protein is thought to be required to maintain the normal checkpoint system in human cell. Loren et al[21] showed that subtle differences in Mad2 protein level markedly altered checkpoint function. Therefore, inactivation of Mad2 would be sufficient to create a haplo-insufficient effect and the loss of mitotic checkpoint control. The most convincing evidence of the role of mitotic checkpoint defect in CIN in mammalian cells obtained in two recent studies showed that disrupting of Mad2 expression resulted in CIN. CIN cells became aneuploidy, a hallmark of cancer associated with aggressive tumor behavior and a poor prognosis[21,22]. Very recently, Poelzl et al[23] reported that estrogen receptor β, which might be involved in the regulation of cellular proliferation, interacted with Mad2 directly and specifically. The Mad2 protein also interacted with the cytoplasmic domain of insulin receptors, which was thought to be a regulator of cellular growth[24,25]. Mad2 protein overexpressed in cancer tissues was exclusively present in the cytoplasm of cancer cells. We speculate that cytoplasmic Mad2 protein may enhance the positive regulatory action of estrogen receptor β and insulin receptor on cellular proliferation. We also discovered that there was a significantly positive correlation between the expressions of Mad2 and p53 (P < 0.05). Orr-Weaver et al[17] thought aneuploidy resulted from defect of Mad2 and other unknown factor could increase the rate at which tumor suppressors, such as p53, were lost. p53 gene was the most frequently mutated gene in human cancer[26-28]. One of its roles was to ensure that when DNA damaged cells were arrested in G1, it attempted to repair their DNA before it was replicated[29,30]. In some cell types, p53 induced apoptosis when overexpressed and was required for apoptosis in response to severe DNA damage[31,32]. p53 might directly activate death genes such as BAX, or down-regulate survival genes such as BCL-2[33-35]. So when p53 gene mutates, aneupoidy from defect of Mad2 might escape from apoptosis and go on its mitosis, finally develop to malignant tumor.

The C/N ratio of Mad2 protein was found to be associated with lymph node metastasis. A C/N ratio greater than 2 was observed more frequently in patients with lymph node metastasis. Therefore, a C/N ratio > 2 may be clinically an important indicator for lymph node metastasis of colorectal cancer.

In conclusion, the protein level of Mad2 is higher in colorectal cancer tissues than in normal tissues. The higher the protein level in colorectal cancer tissues is, the more frequently lymph node metastasis. Since the difference in the protein level between colorectal cancer and normal mucosa is easily detected by immunohistochemistry, Mad2 protein might be a good marker of lymph node metastasis of colorectal cancer.

Footnotes

Edited by Zhu LH and Wang XL

References
1.  Mitelman F, Mertens F, Johansson B. A breakpoint map of recurrent chromosomal rearrangements in human neoplasia. Nat Genet. 1997;15 Spec No:417-474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 496]  [Cited by in F6Publishing: 481]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
2.  Rooney PH, Murray GI, Stevenson DA, Haites NE, Cassidy J, McLeod HL. Comparative genomic hybridization and chromosomal instability in solid tumours. Br J Cancer. 1999;80:862-873.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 100]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
3.  Lengauer C, Kinzler KW, Vogelstein B. Genetic instabilities in human cancers. Nature. 1998;396:643-649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2881]  [Cited by in F6Publishing: 2762]  [Article Influence: 106.2]  [Reference Citation Analysis (0)]
4.  Lengauer C, Kinzler KW, Vogelstein B. Genetic instability in colorectal cancers. Nature. 1997;386:623-627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1367]  [Cited by in F6Publishing: 1338]  [Article Influence: 49.6]  [Reference Citation Analysis (0)]
5.  Lengauer C, Kinzler KW, Vogelstein B. DNA methylation and genetic instability in colorectal cancer cells. Proc Natl Acad Sci USA. 1997;94:2545-2550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 296]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
6.  Li R, Murray AW. Feedback control of mitosis in budding yeast. Cell. 1991;66:519-531.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 853]  [Cited by in F6Publishing: 881]  [Article Influence: 26.7]  [Reference Citation Analysis (0)]
7.  Hoyt MA, Totis L, Roberts BT. S. cerevisiae genes required for cell cycle arrest in response to loss of microtubule function. Cell. 1991;66:507-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 804]  [Cited by in F6Publishing: 831]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
8.  Li Y, Benezra R. Identification of a human mitotic checkpoint gene: hsMAD2. Science. 1996;274:246-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 452]  [Cited by in F6Publishing: 445]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
9.  Taylor SS, McKeon F. Kinetochore localization of murine Bub1 is required for normal mitotic timing and checkpoint response to spindle damage. Cell. 1997;89:727-735.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 412]  [Cited by in F6Publishing: 407]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
10.  Jin DY, Spencer F, Jeang KT. Human T cell leukemia virus type 1 oncoprotein Tax targets the human mitotic checkpoint protein MAD1. Cell. 1998;93:81-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 400]  [Cited by in F6Publishing: 401]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
11.  Barnes DM, Dublin EA, Fisher CJ, Levison DA, Millis RR. Immunohistochemical detection of p53 protein in mammary carcinoma: an important new independent indicator of prognosis. Hum Pathol. 1993;24:469-476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 175]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
12.  Rudner AD, Murray AW. The spindle assembly checkpoint. Curr Opin Cell Biol. 1996;8:773-780.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 261]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
13.  Amon A. The spindle checkpoint. Curr Opin Genet Dev. 1999;9:69-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 277]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
14.  Waters JC, Chen RH, Murray AW, Salmon ED. Localization of Mad2 to kinetochores depends on microtubule attachment, not tension. J Cell Biol. 1998;141:1181-1191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 370]  [Cited by in F6Publishing: 357]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
15.  Sharp-Baker H, Chen RH. Spindle checkpoint protein Bub1 is required for kinetochore localization of Mad1, Mad2, Bub3, and CENP-E, independently of its kinase activity. J Cell Biol. 2001;153:1239-1250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 174]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
16.  Pennisi E. Cell division gatekeepers identified. Science. 1998;279:477-478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
17.  Orr-Weaver TL, Weinberg RA. A checkpoint on the road to cancer. Nature. 1998;392:223-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 93]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
18.  Shah JV, Cleveland DW. Waiting for anaphase: Mad2 and the spindle assembly checkpoint. Cell. 2000;103:997-1000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 234]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
19.  Takahashi T, Haruki N, Nomoto S, Masuda A, Saji S, Osada H, Takahashi T. Identification of frequent impairment of the mitotic checkpoint and molecular analysis of the mitotic checkpoint genes, hsMAD2 and p55CDC, in human lung cancers. Oncogene. 1999;18:4295-4300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 100]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
20.  Chen RH, Brady DM, Smith D, Murray AW, Hardwick KG. The spindle checkpoint of budding yeast depends on a tight complex between the Mad1 and Mad2 proteins. Mol Biol Cell. 1999;10:2607-2618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 143]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
21.  Michel LS, Liberal V, Chatterjee A, Kirchwegger R, Pasche B, Gerald W, Dobles M, Sorger PK, Murty VV, Benezra R. MAD2 haplo-insufficiency causes premature anaphase and chromosome instability in mammalian cells. Nature. 2001;409:355-359.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 574]  [Cited by in F6Publishing: 561]  [Article Influence: 24.4]  [Reference Citation Analysis (0)]
22.  Dobles M, Liberal V, Scott ML, Benezra R, Sorger PK. Chromosome missegregation and apoptosis in mice lacking the mitotic checkpoint protein Mad2. Cell. 2000;101:635-645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 394]  [Cited by in F6Publishing: 388]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
23.  Poelzl G, Kasai Y, Mochizuki N, Shaul PW, Brown M, Mendelsohn ME. Specific association of estrogen receptor beta with the cell cycle spindle assembly checkpoint protein, MAD2. Proc Natl Acad Sci USA. 2000;97:2836-2839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
24.  O'Neill TJ, Zhu Y, Gustafson TA. Interaction of MAD2 with the carboxyl terminus of the insulin receptor but not with the IGFIR. Evidence for release from the insulin receptor after activation. J Biol Chem. 1997;272:10035-10040.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 36]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
25.  Gliozzo B, Sung CK, Scalia P, Papa V, Frasca F, Sciacca L, Giorgino F, Milazzo G, Goldfine ID, Vigneri R. Insulin-stimulated cell growth in insulin receptor substrate-1-deficient ZR-75-1 cells is mediated by a phosphatidylinositol-3-kinase-independent pathway. J Cell Biochem. 1998;70:268-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
26.  Nigro JM, Baker SJ, Preisinger AC, Jessup JM, Hostetter R, Cleary K, Bigner SH, Davidson N, Baylin S, Devilee P. Mutations in the p53 gene occur in diverse human tumour types. Nature. 1989;342:705-708.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1896]  [Cited by in F6Publishing: 1936]  [Article Influence: 55.3]  [Reference Citation Analysis (0)]
27.  Hollstein M, Rice K, Greenblatt MS, Soussi T, Fuchs R, Sørlie T, Hovig E, Smith-Sørensen B, Montesano R, Harris CC. Database of p53 gene somatic mutations in human tumors and cell lines. Nucleic Acids Res. 1994;22:3551-3555.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Greenblatt MS, Bennett WP, Hollstein M, Harris CC. Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res. 1994;54:4855-4878.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Kuerbitz SJ, Plunkett BS, Walsh WV, Kastan MB. Wild-type p53 is a cell cycle checkpoint determinant following irradiation. Proc Natl Acad Sci USA. 1992;89:7491-7495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1145]  [Cited by in F6Publishing: 1256]  [Article Influence: 39.3]  [Reference Citation Analysis (0)]
30.  Nelson WG, Kastan MB. DNA strand breaks: the DNA template alterations that trigger p53-dependent DNA damage response pathways. Mol Cell Biol. 1994;14:1815-1823.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 527]  [Cited by in F6Publishing: 513]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
31.  Lowe SW, Schmitt EM, Smith SW, Osborne BA, Jacks T. p53 is required for radiation-induced apoptosis in mouse thymocytes. Nature. 1993;362:847-849.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1995]  [Cited by in F6Publishing: 1994]  [Article Influence: 64.3]  [Reference Citation Analysis (0)]
32.  Hermeking H, Eick D. Mediation of c-Myc-induced apoptosis by p53. Science. 1994;265:2091-2093.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 525]  [Cited by in F6Publishing: 574]  [Article Influence: 19.1]  [Reference Citation Analysis (0)]
33.  Miyashita T, Krajewski S, Krajewska M, Wang HG, Lin HK, Liebermann DA, Hoffman B, Reed JC. Tumor suppressor p53 is a regulator of bcl-2 and bax gene expression in vitro and in vivo. Oncogene. 1994;9:1799-1805.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Miyashita T, Reed JC. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell. 1995;80:293-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1960]  [Cited by in F6Publishing: 2260]  [Article Influence: 77.9]  [Reference Citation Analysis (0)]
35.  White E. Life, death, and the pursuit of apoptosis. Genes Dev. 1996;10:1-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 968]  [Cited by in F6Publishing: 966]  [Article Influence: 34.5]  [Reference Citation Analysis (0)]