501
|
Herzog CR, Bodon N, Pittman B, Maronpot RR, Massey TE, Anderson MW, You M, Devereux TR. Carcinogen-specific targeting of chromosome 12 for loss of heterozygosity in mouse lung adenocarcinomas: implications for chromosome instability and tumor progression. Oncogene 2004; 23:3033-9. [PMID: 14755239 DOI: 10.1038/sj.onc.1207431] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Genotoxic carcinogens exert their tumorigenic effects in part by inducing genomic instability. We recently showed that loss of heterozygosity (LOH) on chromosome 12 associates significantly with the induction of chromosome instability (CIN) by the likely human lung carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and vinyl carbamate (VC) during mouse lung carcinogenesis. Here, we demonstrate the carcinogen specificity of this event and its effect on lung tumor evolution. LOH on chromosome 12 was observed in 45% of NNK-induced, 59% of VC-induced, 58% of aflatoxin B1 (AFB1)-induced, 14% of N-ethyl-N-nitrosourea (ENU)-induced and 12% of spontaneous lung adenocarcinomas. The frequency of LOH in each of the carcinogen-induced groups, except ENU, was significantly higher than in the spontaneous group (P<0.001). Deletion mapping revealed four potential candidate regions of 1-4 centiMorgans suspected to contain targeted tumor suppressor genes, with at least one expected to have a role in CIN. The relationship between LOH on chromosome 12 and additional chromosomal alterations occurring during lung tumor progression was also examined. LOH on chromosomes 1 and 14 were moderately frequent during malignant progression in tumors from all treatment groups, occurring in 21-35 and 18-33% of tumors. However, these alterations showed significant concurrence with LOH on chromosome 12 in VC-, NNK- and AFB1-induced tumors (P<0.05). The results suggest that a carcinogen-selective mechanism of lung cancer induction involves the frequent inactivation of genes on chromosome 12, including a stability gene that evidently promotes the evolutionary selection of additional chromosomal alterations during malignant progression.
Collapse
|
502
|
Yoshihara T, Ishida M, Kinomura A, Katsura M, Tsuruga T, Tashiro S, Asahara T, Miyagawa K. XRCC3 deficiency results in a defect in recombination and increased endoreduplication in human cells. EMBO J 2004; 23:670-80. [PMID: 14749735 PMCID: PMC1271813 DOI: 10.1038/sj.emboj.7600087] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2003] [Accepted: 12/22/2003] [Indexed: 12/24/2022] Open
Abstract
XRCC3 was inactivated in human cells by gene targeting. Consistent with its role in homologous recombination, XRCC3(-/-) cells showed a two-fold sensitivity to DNA cross-linking agents, a mild reduction in sister chromatid exchange, impaired Rad51 focus formation and elevated chromosome aberrations. Furthermore, endoreduplication was increased five- seven-fold in the mutants. The T241M variant of XRCC3 has been associated with an increased cancer risk. Expression of the wild-type cDNA restored this phenotype, while expression of the variant restored the defective recombinational repair, but not the increased endoreduplication. RPA, a protein essential for homologous recombination and DNA replication, is associated with XRCC3 and Rad52. Overexpression of RPA promoted endoreduplication, which was partially complemented by overexpression of the wild-type XRCC3 protein, but not by overexpression of the variant protein. Overexpression of Rad52 prevented endoreduplication in RPA-overexpressing cells, in XRCC3(-/-) cells and in the variant-expressing cells, suggesting that deregulated RPA was responsible for the increased endoreduplication. These observations offer the first genetic evidence for the association between homologous recombination and replication initiation having a role in cancer susceptibility.
Collapse
Affiliation(s)
- Takashi Yoshihara
- Department of Human Genetics, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
- Department of Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Mari Ishida
- Department of Human Genetics, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Aiko Kinomura
- Department of Human Genetics, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Mari Katsura
- Department of Human Genetics, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Takanori Tsuruga
- Department of Human Genetics, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Satoshi Tashiro
- Department of Biochemistry, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Toshimasa Asahara
- Department of Surgery, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Kiyoshi Miyagawa
- Department of Human Genetics, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
- Department of Human Genetics, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan. Tel.: +81 82 257 5828; Fax: +81 82 256 7102; E-mail:
| |
Collapse
|
503
|
Cotsiki M, Lock RL, Cheng Y, Williams GL, Zhao J, Perera D, Freire R, Entwistle A, Golemis EA, Roberts TM, Jat PS, Gjoerup OV. Simian virus 40 large T antigen targets the spindle assembly checkpoint protein Bub1. Proc Natl Acad Sci U S A 2004; 101:947-52. [PMID: 14732683 PMCID: PMC327122 DOI: 10.1073/pnas.0308006100] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The mitotic spindle checkpoint protein Bub1 has been found to be mutated at low frequency in certain human cancers characterized by aneuploidy. Simian virus 40 large T antigen efficiently immortalizes rodent cells and occasionally transforms them to tumorigenicity. T antigen can also cause genomic instability, inducing chromosomal aberrations and aneuploidy. Here, we report an interaction between Bub1 and T antigen. T antigen coimmunoprecipitates with endogenous Bub1 and Bub3, another component of the spindle checkpoint complex. Genetic analysis demonstrates that the interaction of T antigen with Bub1 is not required for immortalization but is closely correlated with transformation. T antigen induces an override of the spindle checkpoint dependent on Bub1 binding. This interaction with proteins of the spindle checkpoint machinery suggests another role for T antigen and provides insight into its ability to cause chromosomal aberrations, aneuploidy, and transformation.
Collapse
Affiliation(s)
- Marina Cotsiki
- Ludwig Institute for Cancer Research, Royal Free and University College School of Medicine, Courtauld Building, 91 Riding House Street, London W1W 7BS, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
504
|
Dai W, Wang Q, Liu T, Swamy M, Fang Y, Xie S, Mahmood R, Yang YM, Xu M, Rao CV. Slippage of mitotic arrest and enhanced tumor development in mice with BubR1 haploinsufficiency. Cancer Res 2004; 64:440-5. [PMID: 14744753 DOI: 10.1158/0008-5472.can-03-3119] [Citation(s) in RCA: 236] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A compromised spindle checkpoint is thought to play a key role in genetic instability that predisposes cells to malignant transformation. Loss of function mutations of BubR1, an important component of the spindle checkpoint, have been detected in human cancers. Here we show that BubR1(+/-) mouse embryonic fibroblasts are defective in spindle checkpoint activation, contain a significantly reduced amount of securin and Cdc20, and exhibit a greater level of micronuclei than do wild-type cells. RNA interference-mediated down-regulation of BubR1 also greatly reduced securin level. Moreover, compared with wild-type littermates, BubR1(+/-) mice rapidly develop lung as well as intestinal adenocarcinomas in response to challenge with carcinogen. BubR1 is thus essential for spindle checkpoint activation and tumor suppression.
Collapse
Affiliation(s)
- Wei Dai
- Division of Molecular Carcinogenesis, Department of Medicine, New York Medical College, Valhalla, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
505
|
Sihn CR, Suh EJ, Lee KH, Kim TY, Kim SH. p55CDC/hCDC20 mutant induces mitotic catastrophe by inhibiting the MAD2-dependent spindle checkpoint activity in tumor cells. Cancer Lett 2004; 201:203-10. [PMID: 14607335 DOI: 10.1016/s0304-3835(03)00465-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Nondisjunction of chromosomes results in aneuploidy in mammalian cells causing genomic instability. The spindle checkpoint, one of the surveillance systems to maintain genomic stability, prevents missegregation of chromosomes until all the kinetochores are properly attached with bipolar spindles. When this condition is not met, MAD2, a component of the spindle checkpoint complex, associates with p55CDC/hCDC20 to inhibit ubiquitination of substrates by the anaphase-promoting complex (APC). In this study, we have focused on the biological role of the MAD2-binding domain in p55CDC/hCDC20 in the maintenance of genomic stability. Based on previous studies, we constructed a truncated p55CDC/hCDC20 mutant (F2) that harbors only the MAD2-binding domain. Interestingly, we found that in the yeast two-hybrid system, the interaction of F2 and MAD2 was stronger than that of intact p55CDC/hCDC20. We also found that in the presence of the microtubule-disrupting drug, nocodazole, U2OS cells expressing p55CDC/hCDC20 mutants bypassed the mitotic arrest and showed apoptotic morphologies, whereas cells harboring vector alone arrested at metaphase. In particular, the apoptotic phenomena were dramatically enhanced in the F2-expressing cells. These mitotic catastrophes also occurred in cells treated with other microtubule disrupting agents, such as taxol and vinblastine. In addition, the mutant cells exhibited chromosomal missegregation during mitosis, even in the absence of nocodazole. Taken together, these results suggest that agents blocking the spindle checkpoint response may induce tumor cells to become more sensitive to spindle poison drugs, providing a powerful tool to improve chemotherapy.
Collapse
Affiliation(s)
- Choong-Ryoul Sihn
- Department of Biology, Research Institute for Basic Sciences, Kyung Hee University, Hoegi-dong Dongdaemoon-gu, Seoul 130-701, South Korea
| | | | | | | | | |
Collapse
|
506
|
Liu L, Tommasi S, Lee DH, Dammann R, Pfeifer GP. Control of microtubule stability by the RASSF1A tumor suppressor. Oncogene 2003; 22:8125-36. [PMID: 14603253 DOI: 10.1038/sj.onc.1206984] [Citation(s) in RCA: 159] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The RAS association domain family 1A (RASSF1A) gene is silenced by DNA methylation in over 50% of all solid tumors of different histological types. However, the biochemical function of the RASSF1A protein is unknown. We show that RASSF1A colocalizes with microtubules in interphase and decorates spindles and centrosomes during mitosis. RASSF1A has a strong cytoprotective activity against the microtubule-destabilizing drug nocodazole, and against cold-treatment in vivo. Conversely, loss of RASSF1 in RASSF1-/- mouse embryonic fibroblasts renders the cells more sensitive to nocodazole-induced depolymerization of microtubules. The domain required for both microtubule association and stabilization was mapped to a 169 amino-acid fragment that contains the RAS association domain. Overexpression of RASSF1A induces mitotic arrest at metaphase with aberrant mitotic cells reminiscent of such produced by the microtubule-stabilizing drug paclitaxel (taxol), including monopolar spindles, or complete lack of a mitotic spindle. Altered microtubule stability in cells lacking RASSF1A is likely to affect spindle assembly and chromosome attachment, processes that need to be carefully controlled to protect cells from genomic instability and transformation. In addition, knowledge of the microtubule-targeting function of RASSF1 may aid in the development of new anticancer drugs.
Collapse
Affiliation(s)
- Limin Liu
- Division of Biology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | | | | | | | | |
Collapse
|
507
|
Shin HJ, Baek KH, Jeon AH, Park MT, Lee SJ, Kang CM, Lee HS, Yoo SH, Chung DH, Sung YC, McKeon F, Lee CW. Dual roles of human BubR1, a mitotic checkpoint kinase, in the monitoring of chromosomal instability. Cancer Cell 2003; 4:483-97. [PMID: 14706340 DOI: 10.1016/s1535-6108(03)00302-7] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In this study, we show that the formation of polyploidy following sustained mitotic checkpoint activation appears to be preceded by the ubiquitin-dependent proteolysis of hBubR1. In addition, the level of hBubR1 is significantly reduced not only in polyploid cells created by sustained mitotic spindle damage, but also in 21 (31.3%) of 67 human colon adenocarcinomas tested. Importantly, the introduction of hBubR1 triggers the apoptosis of polyploid cells formed by aberrant exit from mitosis and inhibits the growth of tumors established with these cells in athymic nude mice. These results suggest that hBubR1-mediated apoptosis prevents the propagation of cells that breach the mitotic checkpoint and that the control of hBubR1 protein level is an important factor in the acquisition of preneoplastic polyploidy.
Collapse
Affiliation(s)
- Hyun Jin Shin
- Research Institute, National Cancer Center, Goyang, Gyeonggi, 411-764, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
508
|
Christopoulou L, Moore JD, Tyler-Smith C. Over-expression of wild-type Securin leads to aneuploidy in human cells. Cancer Lett 2003; 202:213-8. [PMID: 14643451 DOI: 10.1016/s0304-3835(03)00483-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Changes in gene expression and aneuploidy are both common features of cancers and under- or over-expression of single genes can cause aneuploidy. Securin, which is involved in chromatid separation, has transforming activity in vitro and is over-expressed in many tumours. We have used a GFP-based assay for chromosome loss in human cells to investigate the effects of over-expressing Securin. We find that over-expression causes a two-fold increase in chromosome loss rate and can thus be a direct cause of aneuploidy.
Collapse
Affiliation(s)
- Lina Christopoulou
- Department of Biochemistry, CRC Chromosome Molecular Biology Group, University of Oxford, South Parks Road, OX1 3QU Oxford, UK
| | | | | |
Collapse
|
509
|
Minhas KM, Singh B, Jiang WW, Sidransky D, Califano JA. Spindle assembly checkpoint defects and chromosomal instability in head and neck squamous cell carcinoma. Int J Cancer 2003; 107:46-52. [PMID: 12925955 DOI: 10.1002/ijc.11341] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Alterations in chromosomal number and structure are found in most solid malignancies including head and neck squamous cell carcinoma (HNSC), however, the presence of ongoing, chromosomal instability in HNSC and its relation to spindle assembly checkpoint defects has not been formally demonstrated. We investigated the status of chromosomal instability (CIN) in HNSC primary tumors and cell lines as well as spindle assembly checkpoint integrity in HNSC cell lines. Centromeric fluorescence in situ hybridization (FISH) was carried out on expanded single cell-derived colonies from HNSC cell lines and primary HNSC touch preparations. The deviation of chromosomes from the modal number in single cell derived colonies was 18.4-27% in 6 HNSC cell lines, and 2-3% in a control cell line, HCT116. Twelve primary tumors and 4 normal controls were also studied; all primary tumors demonstrated significant deviation from the modal chromosomal number (average 33.7%, range = 29.9-43.9%), compared to normal controls (average 4.6%, range = 3.6-5.6%). Additional characterization of the rate of chromosomal breakage was carried out by dual color FISH simultaneously using centromeric and telomeric probes for individual chromosomes on expanded singe cell-derived colonies and primary HNSC. Control HCT 116 colonies demonstrated a mean discordance between number of centromeric and telomeric hybridization signals in 21% (range = 19-23%) of cells, whereas HNSC cell line colonies demonstrated a mean discordance of 50% (range = 38-55%), with the majority of instances of discordant signal indicating telomeric loss. Similarly, touch preparations from primary HNSC demonstrated discordance in hybridization signal of centromeric vs. telomeric signal of 26.3% (range = 18.5-42%), with normal controls showing a rate of discordance of 6.4% (range = 4-8%). Finally, all 6 HNSC cell lines demonstrated partial impairment of mitotic arrest in response to nocodazole, indicating that impairment of the spindle assembly checkpoint may contribute to chromosomal instability in HNSC. Ongoing instability in chromosomal number and structure are consistent features of primary HNSC and cell lines. Spindle assembly checkpoint impairment occurs in HNSC cell lines and may contribute to chromosomal instability in HNSC.
Collapse
Affiliation(s)
- Khalid M Minhas
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
510
|
Abstract
p53 regulates a number of genes through transcriptional activation and repression. p53-dependent mitotic checkpoint has been described, but the underlying mechanism is still obscure. Here we examined the effect of p53 on the expression of a human mitotic checkpoint protein, Mitosis Arrest Deficiency 1 (MAD1), in cultured human cells. The expression of MAD1 was reduced when the cells were overexpressing exogenously introduced wild-type p53. The same reduction was also observed when the cells were treated with anticancer agents 5-fluorouracil and cisplatin or were irradiated with UV. Consistently, MAD1 promoter activity diminished in a dose-dependent manner when induced by p53, indicating that p53 repressed MAD1 at a transcriptional level. Intriguingly, several tumor hot spot mutations in p53 (V143A, R175H, R248W, and R273H) did not abolish the ability of p53 to repress MAD1 expression. By serial truncation of the MAD1 promoter, we confined the p53-responsive element to a 38-bp region that represents a novel sequence distinct from the known p53 consensus binding site. Trichostatin A, a histone deacetylase inhibitor, relieved the p53 transrepression activity on MAD1. Chromatin immunoprecipitation assay revealed that p53, histone deacetylase 1, and co-repressor mSin3a associated with the MAD1 promoter in vivo. Taken together, our findings suggest a regulatory mechanism for the mitotic checkpoint in which MAD1 is inhibited by p53.
Collapse
Affiliation(s)
- Abel C S Chun
- Department of Biochemistry, the University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
511
|
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.
Collapse
Affiliation(s)
- Gang-Qiang Li
- Department of Pathology, Anhui Medical University, Hefei, Anhui Province, China.
| | | | | |
Collapse
|
512
|
Rajagopalan H, Nowak MA, Vogelstein B, Lengauer C. The significance of unstable chromosomes in colorectal cancer. Nat Rev Cancer 2003; 3:695-701. [PMID: 12951588 DOI: 10.1038/nrc1165] [Citation(s) in RCA: 332] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A very large fraction of cancers have an abnormal genetic content, called aneuploidy, which is characterized by changes in chromosome structure and number. One explanation for this aneuploidy is chromosomal instability, in which cancer cells gain or lose whole chromosomes or large fractions of chromosomes at a greatly increased rate compared with normal cells. Here, we explore experimental and theoretical evidence for the initiation of chromosomal instability in very early colorectal cancers, and reflect on the role that chromosomal instability could have in colorectal tumorigenesis.
Collapse
Affiliation(s)
- Harith Rajagopalan
- Sidney Kimmel Comprehensive Cancer Center and Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | | | | | | |
Collapse
|
513
|
Weaver BAA, Bonday ZQ, Putkey FR, Kops GJPL, Silk AD, Cleveland DW. Centromere-associated protein-E is essential for the mammalian mitotic checkpoint to prevent aneuploidy due to single chromosome loss. J Cell Biol 2003; 162:551-63. [PMID: 12925705 PMCID: PMC2173788 DOI: 10.1083/jcb.200303167] [Citation(s) in RCA: 200] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Centromere-associated protein-E (CENP-E) is an essential mitotic kinesin that is required for efficient, stable microtubule capture at kinetochores. It also directly binds to BubR1, a kinetochore-associated kinase implicated in the mitotic checkpoint, the major cell cycle control pathway in which unattached kinetochores prevent anaphase onset. Here, we show that single unattached kinetochores depleted of CENP-E cannot block entry into anaphase, resulting in aneuploidy in 25% of divisions in primary mouse fibroblasts in vitro and in 95% of regenerating hepatocytes in vivo. Without CENP-E, diminished levels of BubR1 are recruited to kinetochores and BubR1 kinase activity remains at basal levels. CENP-E binds to and directly stimulates the kinase activity of purified BubR1 in vitro. Thus, CENP-E is required for enhancing recruitment of its binding partner BubR1 to each unattached kinetochore and for stimulating BubR1 kinase activity, implicating it as an essential amplifier of a basal mitotic checkpoint signal.
Collapse
Affiliation(s)
- Beth A A Weaver
- Ludwig Institute for Cancer Research, 3080 CMM-East, 9500 Gilman Drive, La Jolla, CA 92093-0670, USA
| | | | | | | | | | | |
Collapse
|
514
|
Abstract
Chromosome aberrations in human solid tumors are hallmarks of gene deregulation and genome instability. This review summarizes current knowledge regarding aberrations, discusses their functional importance, suggests mechanisms by which aberrations may form during cancer progression and provides examples of clinical advances that have come from studies of chromosome aberrations.
Collapse
Affiliation(s)
- Donna G Albertson
- Cancer Research Institute, University of California San Francisco, San Francisco, California 94143-0808, USA.
| | | | | | | |
Collapse
|
515
|
Toyota M, Sasaki Y, Satoh A, Ogi K, Kikuchi T, Suzuki H, Mita H, Tanaka N, Itoh F, Issa JPJ, Jair KW, Schuebel KE, Imai K, Tokino T. Epigenetic inactivation of CHFR in human tumors. Proc Natl Acad Sci U S A 2003; 100:7818-23. [PMID: 12810945 PMCID: PMC164671 DOI: 10.1073/pnas.1337066100] [Citation(s) in RCA: 146] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2002] [Accepted: 05/08/2003] [Indexed: 01/23/2023] Open
Abstract
Cell-cycle checkpoints controlling the orderly progression through mitosis are frequently disrupted in human cancers. One such checkpoint, entry into metaphase, is regulated by the CHFR gene encoding a protein possessing forkhead-associated and RING finger domains as well as ubiquitin-ligase activity. Although defects in this checkpoint have been described, the molecular basis and prevalence of CHFR inactivation in human tumors are still not fully understood. To address this question, we analyzed the pattern of CHFR expression in a number of human cancer cell lines and primary tumors. We found CpG methylation-dependent silencing of CHFR expression in 45% of cancer cell lines, 40% of primary colorectal cancers, 53% of colorectal adenomas, and 30% of primary head and neck cancers. Expression of CHFR was precisely correlated with both CpG methylation and deacetylation of histones H3 and H4 in the CpG-rich regulatory region. Moreover, CpG methylation and thus silencing of CHFR depended on the activities of two DNA methyltransferases, DNMT1 and DNMT3b, as their genetic inactivation restored CHFR expression. Finally, cells with CHFR methylation had an intrinsically high mitotic index when treated with microtubule inhibitor. This means that cells in which CHFR was epigenetically inactivated constitute loss-of-function alleles for mitotic checkpoint control. Taken together, these findings shed light on a pathway by which mitotic checkpoint is bypassed in cancer cells and suggest that inactivation of checkpoint genes is much more widespread than previously suspected.
Collapse
Affiliation(s)
- Minoru Toyota
- Department of Molecular Biology, Cancer Research Institute, Sapporo Medical University, Sapporo 060-8556, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
516
|
Dobles M, Sorger PK. Mitotic checkpoints, genetic instability, and cancer. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2003; 65:361-8. [PMID: 12760051 DOI: 10.1101/sqb.2000.65.361] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- M Dobles
- Massachusetts Institute of Technology, Department of Biology, Cambridge, Massachusetts 02139, USA
| | | |
Collapse
|
517
|
Abstract
Lung cancer results from multiple changes in the genome of susceptible pulmonary cells caused by exposure to carcinogens found in tobacco smoke, the environment, or the workplace. Recent studies suggest that histologically apparent lung cancer is due to the sequential accumulation of specific genetic and morphologic changes to the normal epithelial cells of the lung. Positive signallers, such as those mediated by the oncogene RAS, and negative signallers, such as those mediated by the tumor suppressor retinoblastoma protein (RB), contribute to unchecked cell growth and proliferation. Other key molecular derangements can also be considered hallmarks of cancer, including evasion of apoptosis and senescence, angiogenesis, tissue invasion, and metastases. Epigenetic inactivation of genes via DNA methylation provides another novel way of evading normal cellular control mechanisms. The new knowledge of the human genome coupled with global methods of detecting genetic abnormalities and profiling gene expression in tumor cells may enable us to understand the signaling pathways of lung cancer cells. These are molecular targets for new cancer therapeutics such as receptor tyrosine kinase inhibitors. This information could advance risk assessment, early detection, prognosis, and therapy for lung cancer.
Collapse
Affiliation(s)
- Yoshitaka Sekido
- Department of Clinical Preventive Medicine, Nagoya University School of Medicine, Tsurumai 65, Showa-ku, Nagoya 466-8560, Japan.
| | | | | |
Collapse
|
518
|
Lee CGL, Ren J, Cheong ISY, Ban KHK, Ooi LLPJ, Yong Tan S, Kan A, Nuchprayoon I, Jin R, Lee KH, Choti M, Lee LA. Expression of the FAT10 gene is highly upregulated in hepatocellular carcinoma and other gastrointestinal and gynecological cancers. Oncogene 2003; 22:2592-603. [PMID: 12730673 DOI: 10.1038/sj.onc.1206337] [Citation(s) in RCA: 149] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The ubiquitin-like modifier (UBL) family has recently generated much interest in the scientific community, as it is implicated to play important regulatory roles via novel protein-protein modification. FAT10 (diubiquitin) belongs to this family of proteins, comprising two ubiquitin-like moieties fused in tandem, and has been implicated to be involved in the maintenance of spindle integrity during mitosis. As FAT10 may play a role in the regulation of genomic stability, we examined if there is an association between FAT10 expression and hepatocellular carcinoma (HCC) or other cancers. Northern blot analyses revealed upregulation of FAT10 expression in the tumors of 90% of HCC patients. In situ hybridization as well as immunohistochemistry utilizing anti-FAT10 antibodies localized highest FAT10 expression in the nucleus of HCC hepatocytes rather than the surrounding immune and non-HCC cells. FAT10 expression was also found to be highly upregulated in other cancers of the gastrointestinal tract and female reproductive system. In conclusion, we demonstrated upregulation of FAT10 expression in various gastrointestinal and gynecological cancers. Its overexpression is unrelated to the general increase in protein synthesis or a general immune/inflammatory response to cancer. Rather, FAT10 may modulate tumorigenesis through its reported interaction with the MAD2 spindle-assembly checkpoint protein.
Collapse
Affiliation(s)
- Caroline G L Lee
- Department of Biochemistry, National University of Singapore, Singapore.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
519
|
Kitagawa K, Abdulle R, Bansal PK, Cagney G, Fields S, Hieter P. Requirement of Skp1-Bub1 interaction for kinetochore-mediated activation of the spindle checkpoint. Mol Cell 2003; 11:1201-13. [PMID: 12769845 DOI: 10.1016/s1097-2765(03)00145-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The spindle checkpoint transiently prevents cell cycle progression of cells that have incurred errors or failed to complete steps during mitosis, including those involving kinetochore function. The molecular nature of the primary signal transmitted from defective kinetochores and how it is detected by the spindle checkpoint are unknown. We report biochemical evidence that Bub1, a component of the spindle checkpoint, associates with centromere (CEN) DNA via Skp1, a core kinetochore component in budding yeast. The Skp1's interaction with Bub1 is required for the mitotic delay induced by kinetochore tension defects, but not for the arrest induced by spindle depolymerization, kinetochore assembly defects, or Mps1 overexpression. We propose that the Skp1-Bub1 interaction is important for transmitting a signal to the spindle checkpoint pathway when insufficient tension is present at kinetochores.
Collapse
Affiliation(s)
- Katsumi Kitagawa
- Department of Molecular Pharmacology, St. Jude Children's Research Hospital, 332 North Lauderdale Street, Memphis, TN 38105, USA.
| | | | | | | | | | | |
Collapse
|
520
|
Yamaguchi H, Aridgides LJ, Zeng W, Osgood C, Young NS, Liu JM. Genetic and transcriptional analysis of spindle checkpoint genes in bone marrow failure patients. Blood Cells Mol Dis 2003; 30:307-11. [PMID: 12737950 DOI: 10.1016/s1079-9796(03)00042-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The evolution of bone marrow failure syndromes such as aplastic anemia (AA) to clonal hematologic diseases such as myelodysplastic syndrome is well recognized. Cytogenetic abnormalities are commonly seen late events, particularly aneuploidy of chromosomes 7 and 8. A proportion of bone marrow failure patients may also develop aneuploidy that is detectable by fluorescence in situ hybridization but not by standard cytogenetic analysis. The molecular basis for aneuploidy in this setting is currently unknown but may include abnormalities in the mitotic spindle checkpoint. For this reason, we searched for mutations in the mitotic spindle checkpoint genes hBUB1 and hMAD2, and also examined the expression of hBUB1 in cells of bone marrow failure patients. No pathogenic mutations were found in 59 patients. Of 170 bone marrow failure patients, less than one-third expressed hBUB1 transcript. Gene expression profiling confirmed a significant down-regulation of hBUB1 message in patients. We conclude that mutations in mitotic spindle checkpoint genes do not account for aneuploidy in marrow failure states. However, we cannot exclude epigenetic inactivation of hBUB1 as a potential mechanism in some patients.
Collapse
Affiliation(s)
- Hiroki Yamaguchi
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
521
|
Nugoli M, Chuchana P, Vendrell J, Orsetti B, Ursule L, Nguyen C, Birnbaum D, Douzery EJP, Cohen P, Theillet C. Genetic variability in MCF-7 sublines: evidence of rapid genomic and RNA expression profile modifications. BMC Cancer 2003; 3:13. [PMID: 12713671 PMCID: PMC156633 DOI: 10.1186/1471-2407-3-13] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2002] [Accepted: 04/24/2003] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Both phenotypic and cytogenetic variability have been reported for clones of breast carcinoma cell lines but have not been comprehensively studied. Despite this, cell lines such as MCF-7 cells are extensively used as model systems. METHODS In this work we documented, using CGH and RNA expression profiles, the genetic variability at the genomic and RNA expression levels of MCF-7 cells of different origins. Eight MCF-7 sublines collected from different sources were studied as well as 3 subclones isolated from one of the sublines by limit dilution. RESULTS MCF-7 sublines showed important differences in copy number alteration (CNA) profiles. Overall numbers of events ranged from 28 to 41. Involved chromosomal regions varied greatly from a subline to another. A total of 62 chromosomal regions were affected by either gains or losses in the 11 sublines studied. We performed a phylogenetic analysis of CGH profiles using maximum parsimony in order to reconstruct the putative filiation of the 11 MCF-7 sublines. The phylogenetic tree obtained showed that the MCF-7 clade was characterized by a restricted set of 8 CNAs and that the most divergent subline occupied the position closest to the common ancestor. Expression profiles of 8 MCF-7 sublines were analyzed along with those of 19 unrelated breast cancer cell lines using home made cDNA arrays comprising 720 genes. Hierarchical clustering analysis of the expression data showed that 7/8 MCF-7 sublines were grouped forming a cluster while the remaining subline clustered with unrelated breast cancer cell lines. These data thus showed that MCF-7 sublines differed at both the genomic and phenotypic levels. CONCLUSIONS The analysis of CGH profiles of the parent subline and its three subclones supported the heteroclonal nature of MCF-7 cells. This strongly suggested that the genetic plasticity of MCF-7 cells was related to their intrinsic capacity to generate clonal heterogeneity. We propose that MCF-7, and possibly the breast tumor it was derived from, evolved in a node like pattern, rather than according to a linear progression model. Due to their capacity to undergo rapid genetic changes MCF-7 cells could represent an interesting model for genetic evolution of breast tumors.
Collapse
Affiliation(s)
- Mélanie Nugoli
- Equipe Génome et Cancer, UMR 5535 CNRS and EMI 0229 INSERM Centre de Recherche CRLC Val d'Aurelle, Montpellier, France
| | - Paul Chuchana
- Equipe Génome et Cancer, UMR 5535 CNRS and EMI 0229 INSERM Centre de Recherche CRLC Val d'Aurelle, Montpellier, France
| | - Julie Vendrell
- Equipe Génome et Cancer, UMR 5535 CNRS and EMI 0229 INSERM Centre de Recherche CRLC Val d'Aurelle, Montpellier, France
| | - Béatrice Orsetti
- Equipe Génome et Cancer, UMR 5535 CNRS and EMI 0229 INSERM Centre de Recherche CRLC Val d'Aurelle, Montpellier, France
| | - Lisa Ursule
- Equipe Génome et Cancer, UMR 5535 CNRS and EMI 0229 INSERM Centre de Recherche CRLC Val d'Aurelle, Montpellier, France
| | - Catherine Nguyen
- Laboratoire TAGC, CIML, Université d'Aix-Marseille II, Marseille, France
| | - Daniel Birnbaum
- INSERM U119 and LBT, Institut Paoli Calmette, 232 blv Ste Marguerite, 13009 Marseille, France
| | - Emmanuel JP Douzery
- Institut des Sciences de l'Evolution de Montpellier CNRS UMR 5554, Université des Sciences et Techniques du Languedoc Montpellier II, Montpellier, France
| | - Pascale Cohen
- Equipe Génome et Cancer, UMR 5535 CNRS and EMI 0229 INSERM Centre de Recherche CRLC Val d'Aurelle, Montpellier, France
- Institut de Biotechnologies et Pharmacologie CNRS UMR 5094, Faculté de Pharmacie Université Montpellier I, Montpellier, France
| | - Charles Theillet
- Equipe Génome et Cancer, UMR 5535 CNRS and EMI 0229 INSERM Centre de Recherche CRLC Val d'Aurelle, Montpellier, France
| |
Collapse
|
522
|
Sashida G, Ito Y, Nakajima A, Kawakubo K, Kuriyama Y, Yagasaki F, Bessho M, Ohyashiki K. Multiple myeloma with monosomy 13 developed in trisomy 13 acute myelocytic leukemia: numerical chromosome abnormality during chromosomal segregation process. CANCER GENETICS AND CYTOGENETICS 2003; 141:154-6. [PMID: 12606134 DOI: 10.1016/s0165-4608(02)00792-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We report here an acute myelocytic leukemia (AML-M2) patient with trisomy 13 as the sole cytogenetic anomaly, who had relapse of AML with a normal karyotype and developed multiple myeloma. Fluorescence in situ hybridization analysis using the RB gene probe revealed the plasma cells of multiple myeloma (MM) to have monosomy 13 anomaly, whereas relapsed blast cells of AML carried disomy of chromosome 13. To our knowledge, this is the first case showing clonal evolution of trisomy 13 AML and monosomy 13 MM, which might be derived from the leukemic clone at relapse.
Collapse
Affiliation(s)
- Goro Sashida
- First Department of Internal Medicine, Tokyo Medical University, Tokyo 160-0023, Japan
| | | | | | | | | | | | | | | |
Collapse
|
523
|
Abstract
The mitotic checkpoint is essential to ensure accurate chromosome segregation by allowing a mitotic delay in response to a spindle defect. This checkpoint postpones the onset of anaphase until all the chromosomes are attached and correctly aligned onto the mitotic spindle. The checkpoint functions by preventing an ubiquitin ligase called the anaphase-promoting complex (APC) from ubiquitinylating proteins whose degradation is required for anaphase onset. Loss of this checkpoint results in chromosome missegregation in higher eukaryotes and may contribute to the genomic instability observed in most of the tumour cells.
Collapse
Affiliation(s)
- Anna Castro
- Centre de recherche de biochimie macromoléculaire, Cnrs UPR 1086, 1919, route de Mende, 34293 Montpellier, France
| | | | | | | |
Collapse
|
524
|
Abstract
Cell-cycle dysregulation is a hallmark of tumor cells. The ability of normal cells to undergo cell-cycle arrest after damage to DNA is crucial for the maintenance of genomic integrity. The biochemical pathways that stop the cell cycle in response to cellular stressors are called checkpoints. Defective checkpoint function results in genetic modifications that contribute to tumorigenesis. The regulation of checkpoint signaling also has important clinical implications because the abrogation of checkpoint function can alter the sensitivity of tumor cells to chemotherapeutics. Here, we provide an overview of the mechanisms that regulate the cell cycle, current anticancer therapies that target checkpoint signaling pathways, and strategies for the development of novel chemotherapeutic agents.
Collapse
Affiliation(s)
- Zoe A Stewart
- Department of Biochemistry, Center in Molecular Toxicology and the Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | |
Collapse
|
525
|
Cleveland DW, Mao Y, Sullivan KF. Centromeres and kinetochores: from epigenetics to mitotic checkpoint signaling. Cell 2003; 112:407-21. [PMID: 12600307 DOI: 10.1016/s0092-8674(03)00115-6] [Citation(s) in RCA: 788] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The centromere is a chromosomal locus that ensures delivery of one copy of each chromosome to each daughter at cell division. Efforts to understand the nature and specification of the centromere have demonstrated that this central element for ensuring inheritance is itself epigenetically determined. The kinetochore, the protein complex assembled at each centromere, serves as the attachment site for spindle microtubules and the site at which motors generate forces to power chromosome movement. Unattached kinetochores are also the signal generators for the mitotic checkpoint, which arrests mitosis until all kinetochores have correctly attached to spindle microtubules, thereby representing the major cell cycle control mechanism protecting against loss of a chromosome (aneuploidy).
Collapse
Affiliation(s)
- Don W Cleveland
- Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | | | | |
Collapse
|
526
|
Wassmann K, Liberal V, Benezra R. Mad2 phosphorylation regulates its association with Mad1 and the APC/C. EMBO J 2003; 22:797-806. [PMID: 12574116 PMCID: PMC145433 DOI: 10.1093/emboj/cdg071] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Improper attachment of the mitotic spindle to the kinetochores of paired sister chromatids in mitosis is monitored by a checkpoint that leads to an arrest in early metaphase. This arrest requires the inhibitory association of Mad2 with the anaphase promoting complex/cyclosome (APC/C). It is not known how the association of Mad2 with the kinetochore and the APC/C is regulated in mitosis. Here, we demonstrate that human Mad2 is modified through phosphorylation on multiple serine residues in vivo in a cell cycle dependent manner and that only unphosphorylated Mad2 interacts with Mad1 or the APC/C in vivo. A Mad2 mutant containing serine to aspartic acid mutations mimicking the C-terminal phosphorylation events fails to interact with Mad1 or the APC/C and acts as a dominant-negative antagonist of wild-type Mad2. These data suggest that the phosphorylation state of Mad2 regulates its checkpoint activity by modulating its association with Mad1 and the APC/C.
Collapse
Affiliation(s)
- Katja Wassmann
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, Box 241, 1275 York Avenue, 10021 New York, NY, USA and Laboratoire de Biologie Cellulaire du Développement, UMR7622, CNRS, Université Pierre et Marie Curie, 9 quai Saint Bernard, 75005 Paris, France Corresponding author e-mail:
| | - Vasco Liberal
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, Box 241, 1275 York Avenue, 10021 New York, NY, USA and Laboratoire de Biologie Cellulaire du Développement, UMR7622, CNRS, Université Pierre et Marie Curie, 9 quai Saint Bernard, 75005 Paris, France Corresponding author e-mail:
| | - Robert Benezra
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, Box 241, 1275 York Avenue, 10021 New York, NY, USA and Laboratoire de Biologie Cellulaire du Développement, UMR7622, CNRS, Université Pierre et Marie Curie, 9 quai Saint Bernard, 75005 Paris, France Corresponding author e-mail:
| |
Collapse
|
527
|
Babu JR, Jeganathan KB, Baker DJ, Wu X, Kang-Decker N, van Deursen JM. Rae1 is an essential mitotic checkpoint regulator that cooperates with Bub3 to prevent chromosome missegregation. J Cell Biol 2003; 160:341-53. [PMID: 12551952 PMCID: PMC2172680 DOI: 10.1083/jcb.200211048] [Citation(s) in RCA: 279] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The WD-repeat proteins Rae1 and Bub3 show extensive sequence homology, indicative of functional similarity. However, previous studies have suggested that Rae1 is involved in the mRNA export pathway and Bub3 in the mitotic checkpoint. To determine the in vivo roles of Rae1 and Bub3 in mammals, we generated knockout mice that have these genes deleted individually or in combination. Here we show that haplo-insufficiency of either Rae1 or Bub3 results in a similar phenotype involving mitotic checkpoint defects and chromosome missegregation. We also show that overexpression of Rae1 can correct for Rae1 haplo-insufficiency and, surprisingly, Bub3 haplo-insufficiency. Rae1-null and Bub3-null mice are embryonic lethal, although cells from these mice did not have a detectable defect in nuclear export of mRNA. Unlike null mice, compound haplo-insufficient Rae1/Bub3 mice are viable. However, cells from these mice exhibit much greater rates of premature sister chromatid separation and chromosome missegregation than single haplo-insufficient cells. Finally, we show that mice with mitotic checkpoint defects are more susceptible to dimethylbenzanthrene-induced tumorigenesis than wild-type mice. Thus, our data demonstrate a novel function for Rae1 and characterize Rae1 and Bub3 as related proteins with essential, overlapping, and cooperating roles in the mitotic checkpoint.
Collapse
Affiliation(s)
- J Ramesh Babu
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | |
Collapse
|
528
|
Blanchard JM. [Oncogenes and mitotic regulators: a change in perspective of our view of neoplastic processes]. Med Sci (Paris) 2003; 19:187-99. [PMID: 12836613 DOI: 10.1051/medsci/2003192187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Our vision of the cancer cell has dramatically changed since the discovery of proto-oncogenes, whose deregulation was proposed to mimic normal growth signalling. This notion, linking cancer to cell signalling pathways, has progressively led the way to the concept of the mutator phenotype, in which genetic instability plays an essential role in the onset of cancer. This then transformed cancer into a DNA repair disease. However, as foreseen decades ago by cytogeneticists, point mutations are not sufficient to give a full picture of the whole process. As a result, aneuploidy, rather than gene mutation, has been proposed as the explanation for the complex changes observed in cancer cells. The culprits were found among genes involved in the control of the cell division cycle, and work aimed at understanding the regulation of S phase and mitosis have yielded new insights into our understanding of cancer.
Collapse
Affiliation(s)
- Jean-Marie Blanchard
- Institut de Génétique Moléculaire, Cnrs UMR 5535, IFR24, Equipe labellisée La Ligue, 1919, route de Mende, 34293 Montpellier, France.
| |
Collapse
|
529
|
Liu ST, van Deursen JM, Yen TJ. The role of mitotic checkpoint in maintaining genomic stability. Curr Top Dev Biol 2003; 58:27-51. [PMID: 14711012 DOI: 10.1016/s0070-2153(03)58002-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Song-Tao Liu
- Fox Chase Cancer Center, 7701 Burholme Avenue, Philadelphia, PA 19111, USA
| | | | | |
Collapse
|
530
|
Kitagawa R, Law E, Tang L, Rose AM. The Cdc20 homolog, FZY-1, and its interacting protein, IFY-1, are required for proper chromosome segregation in Caenorhabditis elegans. Curr Biol 2002; 12:2118-23. [PMID: 12498686 DOI: 10.1016/s0960-9822(02)01392-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Accurate chromosome segregation is achieved by a series of highly regulated processes that culminate in the metaphase-to-anaphase transition of the cell cycle. In the budding yeast Saccharomyces cerevisiae, the degradation of the securin protein Pds1 reverses the binding and inhibition of the separase protein Esp1. Esp1 cleaves Scc1. That cleavage promotes the dissociation of the cohesin complex from the chromosomes and leads the separation of sister chromatids. Proteolysis of Pds1 is regulated by the anaphase-promoting complex (APC), a large multi-subunit E3 ubiquitin ligase whose activity is regulated by Cdc20/Fizzy. We have previously shown that the Caenorhabditis elegans genes mdf-1/MAD1 and mdf-2/MAD2 encode key members of the spindle checkpoint. Loss of function of either gene leads to an accumulation of somatic and heritable defects and ultimately results in death. Here we show that a missense mutation in fzy-1/CDC20/Fizzy suppresses mdf-1 lethality. We identified a FZY-1-interacting protein, IFY-1, a novel destruction-box protein. IFY-1 accumulates in one-cell-arrested emb-30/APC4 embryos and interacts with SEP-1, a C. elegans separase, suggesting that IFY-1 functions as a C. elegans securin.
Collapse
Affiliation(s)
- Risa Kitagawa
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada.
| | | | | | | |
Collapse
|
531
|
Habu T, Kim SH, Weinstein J, Matsumoto T. Identification of a MAD2-binding protein, CMT2, and its role in mitosis. EMBO J 2002; 21:6419-28. [PMID: 12456649 PMCID: PMC136962 DOI: 10.1093/emboj/cdf659] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
MAD2 is a key component of the spindle checkpoint that delays the onset of anaphase until all the kinetochores are attached to the spindle. It binds to human p55CDC and prevents it from promoting destruction of an anaphase inhibitor, securin. Here we report the characterization of a novel MAD2-binding protein, CMT2. Upon the completion of spindle attachment, formation of the CMT2-MAD2 complex coincides with dissociation of the p55CDC-MAD2 complex. Overexpression of CMT2 in cells arrested by the spindle checkpoint causes premature destruction of securin and allows exit from mitosis without chromosome segregation. Depletion of CMT2 induces cell death following a transient delay in the onset of anaphase. These results indicate that CMT2 interacts with the spindle checkpoint and coordinates cell cycle events in late mitosis.
Collapse
Affiliation(s)
- Toshiyuki Habu
- Departments of Radiation Oncology and Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, Amgen Inc., 5-2-A, One Amgen Center Drive, Thousand Oaks, CA 91320, USA and Radiation Biology Center, Kyoto University, Yoshida-Konoe cho, Sakyo ku, Kyoto, Japan Present address: Radiation Biology Center, Kyoto University, Yoshida-Konoe cho, Sakyo ku, Kyoto, Japan Present address: Department of Biology, Kyung Hee University, Seoul, 130-701, Republic of Korea Corresponding author e-mail:
| | - Sang Hoon Kim
- Departments of Radiation Oncology and Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, Amgen Inc., 5-2-A, One Amgen Center Drive, Thousand Oaks, CA 91320, USA and Radiation Biology Center, Kyoto University, Yoshida-Konoe cho, Sakyo ku, Kyoto, Japan Present address: Radiation Biology Center, Kyoto University, Yoshida-Konoe cho, Sakyo ku, Kyoto, Japan Present address: Department of Biology, Kyung Hee University, Seoul, 130-701, Republic of Korea Corresponding author e-mail:
| | - Jasminder Weinstein
- Departments of Radiation Oncology and Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, Amgen Inc., 5-2-A, One Amgen Center Drive, Thousand Oaks, CA 91320, USA and Radiation Biology Center, Kyoto University, Yoshida-Konoe cho, Sakyo ku, Kyoto, Japan Present address: Radiation Biology Center, Kyoto University, Yoshida-Konoe cho, Sakyo ku, Kyoto, Japan Present address: Department of Biology, Kyung Hee University, Seoul, 130-701, Republic of Korea Corresponding author e-mail:
| | - Tomohiro Matsumoto
- Departments of Radiation Oncology and Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, Amgen Inc., 5-2-A, One Amgen Center Drive, Thousand Oaks, CA 91320, USA and Radiation Biology Center, Kyoto University, Yoshida-Konoe cho, Sakyo ku, Kyoto, Japan Present address: Radiation Biology Center, Kyoto University, Yoshida-Konoe cho, Sakyo ku, Kyoto, Japan Present address: Department of Biology, Kyung Hee University, Seoul, 130-701, Republic of Korea Corresponding author e-mail:
| |
Collapse
|
532
|
Kuramoto K, Ban S, Oda K, Tanaka H, Kimura A, Suzuki G. Chromosomal instability and radiosensitivity in myelodysplastic syndrome cells. Leukemia 2002; 16:2253-8. [PMID: 12399970 DOI: 10.1038/sj.leu.2402703] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2002] [Accepted: 06/20/2002] [Indexed: 11/08/2022]
Abstract
Myelodysplastic syndrome (MDS) is a clonal disorder of hematopoietic stem cells. To investigate whether chromosomal instability and/or DNA repair defects are involved in the development of MDS, we measured the micronucleus (MN) frequency in peripheral blood lymphocytes exposed to various doses of X-rays, using a cytokinesis-block micronucleus assay. The spontaneous MN frequencies in RAEB and RAEB-T patients were significantly higher than those in normal individuals (P = 0.0224, P = 0.008, respectively). Also, the X-ray-induced MN frequencies in RA/RARS, RAEB, and RAEB-T patients were significantly higher than those in normal individuals (P = 0.007, P = 0.003, P = 0.003, respectively, at 2 Gy). In order to elucidate the cause of unusual radiosensitivity, we measured the expression levels of nucleotide excision repair (NER) genes in peripheral blood mononuclear cells using a RT-PCR method. Reduction of NER gene expression was found in only one of 10 patients with low risk MDS, but in four of 11 patients with high risk MDS. Our data suggest that chromosomal instability and DNA repair defects may be involved in the pathophysiology of disease progression of MDS.
Collapse
Affiliation(s)
- K Kuramoto
- Department of Clinical Studies, Radiation Effects Research Foundation, Hiroshima, Japan
| | | | | | | | | | | |
Collapse
|
533
|
Plug-Demaggio AW, McDougall JK. The human papillomavirus type 16 E6 oncogene induces premature mitotic chromosome segregation. Oncogene 2002; 21:7507-13. [PMID: 12386813 DOI: 10.1038/sj.onc.1205903] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2002] [Revised: 07/12/2002] [Accepted: 07/18/2002] [Indexed: 11/09/2022]
Abstract
Expression of the human papillomavirus type 16 E6 and E7 oncogenes initiates and maintains abnormal cell replication, by interacting with the p53 and retinoblastoma (Rb) gene products. Subsequent changes in host cell gene expression, as a consequence of genetic instability, can result in progression to invasive carcinoma. In addition to previously described effects of these viral oncogenes on centrosome synthesis, primarily associated with the expression of E7, the results described herein demonstrate that the E6 oncogene can induce premature chromosome segregation in human cells.
Collapse
Affiliation(s)
- Annemieke W Plug-Demaggio
- Department of Pathology, University of Washington, 1959 Pacific Street, N.E., Seattle, Washington, WA 98195-7407, USA
| | | |
Collapse
|
534
|
Bahar R, O-Wang J, Kawamura K, Seimiya M, Wang Y, Hatano M, Okada S, Tokuhisa T, Watanabe T, Tagawa M. Growth retardation, polyploidy, and multinucleation induced by Clast3, a novel cell cycle-regulated protein. J Biol Chem 2002; 277:40012-40019. [PMID: 12147697 DOI: 10.1074/jbc.m205345200] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have identified a novel gene, Clast3, by subtraction of cDNAs derived from activated and naive B lymphocytes. Clast3 expression is elevated in cycling cells and down-regulated in cells undergoing growth arrest, indicating that its expression is controlled in a cell cycle-dependent manner. The deduced amino acid sequence of Clast3 cDNA exhibits no significant homology to the known proteins in mammalian and other species. Immunofluorescence staining revealed that Clast3 localizes into discrete nuclear foci. Forced expression of Clast3 results in growth retardation, polyploidy, and generation of multinucleated cells. Treatment of Clast3 transfectants with nocodazole, a spindle-damaging agent, greatly enhances the incidence of the multinucleated cells, suggesting that Clast3 overexpression impairs the same checkpoint activated by nocodazole. Down-regulation of Clast3 expression by antisense oligonucleotides results in a decrease of cells at G(2)-M phase and a concomitant increase of apoptotic cells. These findings indicate that Clast3 is a novel cell cycle-regulated protein and that its constitutive overexpression induces polyploidy and multinucleation by interfering with the mitotic spindle checkpoint.
Collapse
Affiliation(s)
- Rumana Bahar
- Division of Pathology, Chiba Cancer Center Research Institute, Chiba 260-8717, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
535
|
Masuda A, Takahashi T. Chromosome instability in human lung cancers: possible underlying mechanisms and potential consequences in the pathogenesis. Oncogene 2002; 21:6884-97. [PMID: 12362271 DOI: 10.1038/sj.onc.1205566] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Chromosomal abnormality is one of the hallmarks of neoplastic cells, and the persistent presence of chromosome instability (CIN) has been demonstrated in human cancers, including lung cancer. Recent progress in molecular and cellular biology as well as cytogenetics has shed light on the underlying mechanisms and the biological and clinical significance of chromosome abnormalities and the CIN phenotype. Chromosome abnormalities can be classified broadly into numerical (i.e., aneuploidy) and structural alterations (e.g., deletion, translocation, homogenously staining region (HSR), double minutes (DMs)). However, both alterations usually occur in the same cells, suggesting some overlap in their underlying mechanisms. Missegregation of chromosomes may result from various causes, including defects of mitotic spindle checkpoint, abnormal centrosome formation and failure of cytokinesis, while structural alterations of chromosomes may be caused especially by failure in the repair of DNA double-strand breaks (DSBs) due to the impairment of DNA damage checkpoints and/or DSB repair systems. Recent studies also suggest that telomere erosion may be involved. The consequential acquisition of the CIN phenotype would give lung cancer cells an excellent opportunity to efficiently alter their characteristics so as to be more malignant and suitable to their microenvironment, thereby gaining a selective growth advantage.
Collapse
Affiliation(s)
- Akira Masuda
- Division of Molecular Oncology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan
| | | |
Collapse
|
536
|
Musacchio A, Hardwick KG. The spindle checkpoint: structural insights into dynamic signalling. Nat Rev Mol Cell Biol 2002; 3:731-41. [PMID: 12360190 DOI: 10.1038/nrm929] [Citation(s) in RCA: 409] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chromosome segregation is a complex and astonishingly accurate process whose inner working is beginning to be understood at the molecular level. The spindle checkpoint plays a key role in ensuring the fidelity of this process. It monitors the interactions between chromosomes and microtubules, and delays mitotic progression to allow extra time to correct defects. Here, we review and integrate findings on the dynamics of checkpoint proteins at kinetochores with structural information about signalling complexes.
Collapse
Affiliation(s)
- Andrea Musacchio
- Department of Experimental Oncology, European Institute of Oncology, Via Ripamonti 435, 20141 Milan, Italy.
| | | |
Collapse
|
537
|
Reith A, Sudbø J. Impact of genomic instability in risk assessment and chemoprevention of oral premalignancies. Int J Cancer 2002; 101:205-9. [PMID: 12209969 DOI: 10.1002/ijc.10569] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Head-and-neck cancer is a disfiguring disease with increasing incidence rates even in young people, whose exposure to known risk factors is limited. This emphasizes the importance of early identification, on an individual basis, of precursor lesions that will develop into carcinomas. The clinical value of identifying individuals at high risk of oral cancer is emphasized by the fact that these patients are likely to benefit from available chemopreventive measures, largely without adverse effects.
Collapse
Affiliation(s)
- Albrecht Reith
- Department of Pathology, Norwegian Radium Hospital, University of Oslo, Norway
| | | |
Collapse
|
538
|
Abstract
Faithful transmission of chromosomes during mitosis is ensured by the spindle assembly checkpoint. This molecular safeguard examines whether prerequisites for chromosome segregation have been satisfied and thereby determines whether to execute or to delay chromosome segregation. Only when all the chromosomes are attached by kinetochore microtubules from two opposite spindle poles and proper tension is placed on the paired kinetochores does anaphase take place, allowing the physical splitting of sister chromatids. Recent studies have provided novel insights into the molecular mechanisms through which the spindle assembly checkpoint is regulated by both the attachment of chromosomes to kinetochore microtubules and the tension exerted on kinetochores.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
539
|
Harper JW, Burton JL, Solomon MJ. The anaphase-promoting complex: it's not just for mitosis any more. Genes Dev 2002; 16:2179-206. [PMID: 12208841 DOI: 10.1101/gad.1013102] [Citation(s) in RCA: 372] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- J Wade Harper
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
540
|
Putkey FR, Cramer T, Morphew MK, Silk AD, Johnson RS, McIntosh JR, Cleveland DW. Unstable kinetochore-microtubule capture and chromosomal instability following deletion of CENP-E. Dev Cell 2002; 3:351-65. [PMID: 12361599 DOI: 10.1016/s1534-5807(02)00255-1] [Citation(s) in RCA: 243] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
A selective disruption of the mouse CENP-E gene was generated to test how this kinetochore-associated, kinesin-like protein contributes to chromosome segregation. The removal of CENP-E in primary cells produced spindles in which some metaphase chromosomes lay juxtaposed to a spindle pole, despite the absence of microtubules stably bound to their kinetochores. Most CENP-E-free chromosomes moved to the spindle equator, but their kinetochores bound only half the normal number of microtubules. Deletion of CENP-E in embryos led to early developmental arrest. Selective deletion of CENP-E in liver revealed that tissue regeneration after chemical damage was accompanied by aberrant mitoses marked by chromosome missegregation. CENP-E is thus essential for the maintenance of chromosomal stability through efficient stabilization of microtubule capture at kinetochores.
Collapse
Affiliation(s)
- Frances R Putkey
- Ludwig Institute for Cancer Research, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | | | | | | | | | |
Collapse
|
541
|
Lentini L, Pipitone L, Di Leonardo A. Functional inactivation of pRB results in aneuploid mammalian cells after release from a mitotic block. Neoplasia 2002; 4:380-7. [PMID: 12192596 PMCID: PMC1661677 DOI: 10.1038/sj.neo.7900256] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2002] [Accepted: 04/15/2002] [Indexed: 11/08/2022]
Abstract
The widespread chromosome instability observed in tumors and in early stage carcinomas suggests that aneuploidy could be a prerequisite for cellular transformation and tumor initiation. Defects in tumor suppressors and genes that are part of mitotic checkpoints are likely candidates for the aneuploid phenotype. By using flow cytometric, cytogenetic, and immunocytochemistry techniques we investigated whether pRB deficiency could drive perpetual aneuploidy in normal human and mouse fibroblasts after mitotic checkpoint challenge by microtubule-destabilizing drugs. Both mouse and human pRB-deficient primary fibroblasts resulted, upon release from a mitotic block, in proliferating aneuploid cells possessing supernumerary centrosomes. Aneuploid pRB-deficient cells show an elevated variation in chromosome numbers among cells of the same clone. In addition, these cells acquired the capability to grow in an anchorage-independent way at the same extent as tumor cells did suggesting aneuploidy as an initial mutational step in cell transformation. Normal Mouse Embryonic Fibroblasts (MEFs) harboring LoxP sites flanking exon 19 of the Rb gene arrested in G2/M with duplicated centrosomes after colcemid treatment. However, these cells escaped the arrest and became aneuploid upon pRB ablation by CRE recombinase, suggesting pRB as a major component of a checkpoint that controls cellular ploidy.
Collapse
Affiliation(s)
- Laura Lentini
- Department of Cell and Developmental Biology A. Monroy, University of Palermo, viale delle Scienze 90128, Palermo, Italy
| | | | | |
Collapse
|
542
|
Bouffler S, Silver A, Cox R. Mechanistic and genetic studies of radiation tumorigenesis in the mouse--implications for low dose risk estimation. JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2002; 22:A11-A16. [PMID: 12400940 DOI: 10.1088/0952-4746/22/3a/302] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Radiation cancer risk estimates remain firmly based upon epidemiological data. Experimental validation of the fundamental aspects of these risk estimates relies on animal studies. In particular, animal model systems for radiation carcinogenesis can provide data for mechanistic modelling approaches to risk estimation. The accuracy and validity of risk estimation models developed will depend upon the extent of our understanding of the process of radiation carcinogenesis. The study of 'spontaneous' tumours in humans continues to provide a sound context in which to consider the mechanisms of radiation carcinogenesis. Several mouse radiation carcinogenesis systems are considered here with particular reference to the nature of the initiating event and the influence of genetic susceptibility on radiation-induced cancer.
Collapse
Affiliation(s)
- Simon Bouffler
- National Radiological Protection Board, Chilton, Didcot, Oxfordshire, UK
| | | | | |
Collapse
|
543
|
Blajeski AL, Phan VA, Kottke TJ, Kaufmann SH. G(1) and G(2) cell-cycle arrest following microtubule depolymerization in human breast cancer cells. J Clin Invest 2002; 110:91-9. [PMID: 12093892 PMCID: PMC151025 DOI: 10.1172/jci13275] [Citation(s) in RCA: 168] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Microtubule-depolymerizing agents are widely used to synchronize cells, screen for mitotic checkpoint defects, and treat cancer. The present study evaluated the effects of these agents on normal and malignant human breast cell lines. After treatment with 1 microM nocodazole, seven of ten breast cancer lines (type A cells) arrested in mitosis, whereas the other three (type B cells) did not. Similar effects were observed with 100 nM vincristine or colchicine. Among five normal mammary epithelial isolates, four exhibited type A behavior and one exhibited type B behavior. Further experiments revealed that the type B cells exhibited a biphasic dose-response curve, with mitotic arrest at low drug concentrations (100 nM nocodazole or 6 nM vincristine) that failed to depolymerize microtubules and a p53-independent p21(waf1/cip1)-associated G(1) and G(2) arrest at higher concentrations (1 microM nocodazole or 100 nM vincristine) that depolymerized microtubules. Collectively, these observations provide evidence for coupling of premitotic cell-cycle progression to microtubule integrity in some breast cancer cell lines (representing a possible "microtubule integrity checkpoint") and suggest a potential explanation for the recently reported failure of some cancer cell lines to undergo nocodazole-induced mitotic arrest despite intact mitotic checkpoint proteins.
Collapse
Affiliation(s)
- April L Blajeski
- Department of Molecular Pharmacology and Experimental Therapeutics, Tumor Biology Program, Mayo Graduate School, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
544
|
Abstract
Complex living organisms possess qualities that cannot be reduced to the simple addition of quantities. Among such qualities are a specific form and a specific organization. Thinking about morphological aspects is a prime example of the qualitative approach to biological matters. Such a morphogenetic perspective has been continuously developed, both theoretically and experimentally, along the past century, even though it is now rather marginal within a mainstream dominated by molecular biology. However, the morphogenetic outlook can be applied to the understanding of complex biological phenomena, such as cancer. This phenomenon is currently explained as a cellular problem caused by specific gene mutations and/or specific loss of gene regulation. Nevertheless, cancer is a problem that affects the whole organism. Contemporary research based on the genetic paradigm of cancer causation has led to paradoxes and anomalies that cannot be explained within such a reductionist paradigm. Here it is proposed that real, non-experimental, sporadic cancer may be understood as a conflict between an organized morphology (the organism) and a part of such a morphology that drifts towards an amorphous state (the tumour). Thus, rare, sporadic cancer in children can be the result of early disruption of the developmental constraints before the organism has achieved its morphological maturity. While common sporadic cancer in aged individuals may ensue as a result of the weakening or exhaustion of the developmental constraints that determine the morphological stability of the organism, once the organism is past its reproductive prime.
Collapse
Affiliation(s)
- A Aranda-Anzaldo
- Laboratorio de Biología Molecular, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Mexico.
| |
Collapse
|
545
|
Bai C, Ramanan N, Wang YM, Wang Y. Spindle assembly checkpoint component CaMad2p is indispensable for Candida albicans survival and virulence in mice. Mol Microbiol 2002; 45:31-44. [PMID: 12100546 DOI: 10.1046/j.1365-2958.2002.02995.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Here, we report an indispensable role for spindle assembly checkpoint (SAC) component CaMad2p in the survival and virulence of Candida albicans in mice. We hypothesized that cell cycle checkpoint functions, especially those monitoring the integrity of DNA and chromosome segregation, might be required for the pathogen to repair damage caused by host defence. To test this idea, we created SAC-defective mutants by deleting the CaMAD2 gene that encodes a key component of the SAC pathway. The CaMAD2 mutant appears normal in morphology, growth rate and growth mode switch in unperturbed conditions. However, it quickly loses viability when treated with nocodazole, which causes disassembly of mitotic spindles. The mutant also exhibits increased frequency of chromosome loss. The virulence of the mutant is greatly reduced in mice, presumably because of the inability of the mutant cells to stop the cell cycle when the host defence damages cellular components important for chromosome segregation. Supporting this hypothesis, unlike the wild-type cells that can proliferate within and eventually grow out of macrophages, most of the CaMAD2 null mutant cells are unable to survive. This study suggests that SAC is required for survival of C. albicans in the host and could thus be targeted for anti-C. albicans therapies.
Collapse
Affiliation(s)
- Chen Bai
- Microbial Collection and Screening Laboratory, Institute of Molecular and Cell Biology, Singapore
| | | | | | | |
Collapse
|
546
|
Hagting A, Den Elzen N, Vodermaier HC, Waizenegger IC, Peters JM, Pines J. Human securin proteolysis is controlled by the spindle checkpoint and reveals when the APC/C switches from activation by Cdc20 to Cdh1. J Cell Biol 2002; 157:1125-37. [PMID: 12070128 PMCID: PMC2173548 DOI: 10.1083/jcb.200111001] [Citation(s) in RCA: 247] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Progress through mitosis is controlled by the sequential destruction of key regulators including the mitotic cyclins and securin, an inhibitor of anaphase whose destruction is required for sister chromatid separation. Here we have used live cell imaging to determine the exact time when human securin is degraded in mitosis. We show that the timing of securin destruction is set by the spindle checkpoint; securin destruction begins at metaphase once the checkpoint is satisfied. Furthermore, reimposing the checkpoint rapidly inactivates securin destruction. Thus, securin and cyclin B1 destruction have very similar properties. Moreover, we find that both cyclin B1 and securin have to be degraded before sister chromatids can separate. A mutant form of securin that lacks its destruction box (D-box) is still degraded in mitosis, but now this is in anaphase. This destruction requires a KEN box in the NH2 terminus of securin and may indicate the time in mitosis when ubiquitination switches from APCCdc20 to APCCdh1. Lastly, a D-box mutant of securin that cannot be degraded in metaphase inhibits sister chromatid separation, generating a cut phenotype where one cell can inherit both copies of the genome. Thus, defects in securin destruction alter chromosome segregation and may be relevant to the development of aneuploidy in cancer.
Collapse
Affiliation(s)
- Anja Hagting
- Wellcome/Cancer Research UK Institute, Cambridge CB2 1QR, United Kingdom
| | | | | | | | | | | |
Collapse
|
547
|
Herzig M, Christofori G. Recent advances in cancer research: mouse models of tumorigenesis. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1602:97-113. [PMID: 12020798 DOI: 10.1016/s0304-419x(02)00039-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Over the past 20 years, cancer research has gained major insights into the complexity of tumor development, in particular into the molecular mechanisms that underlie the progressive transformation of normal cells into highly malignant derivatives. It is estimated that the transformation of a normal cell to a malignant tumor cell is dependent upon a small number of genetic alterations, estimated to be within the range of four to seven rate-limiting events. Critical events in the evolution of neoplastic disease include the loss of proliferative control, the failure to undergo programmed cell death (apoptosis), the onset of neoangiogenesis, tissue remodeling, invasion of tumor cells into surrounding tissue and, finally, metastatic dissemination of tumor cells to distant organs. In patients, the molecular analysis of these multiple steps is hampered by the unavailability of tumor biopsies from all tumor stages. In contrast, mouse models of tumorigenesis allow the reproducible isolation of all tumor stages, including normal tissue, which are then amenable to pathological, genetic and biochemical analyses and, hence, have been instrumental in investigating cancer-related genes and their role in carcinogenesis. In this review, we discuss mouse tumor models that have contributed substantially to the identification and characterization of novel tumor pathways. In particular, we focus on transgenic and knockout mouse models that closely mimic human cancer and thus can be used as model systems for cancer research.
Collapse
|
548
|
Canman JC, Salmon ED, Fang G. Inducing precocious anaphase in cultured mammalian cells. CELL MOTILITY AND THE CYTOSKELETON 2002; 52:61-5. [PMID: 12112148 DOI: 10.1002/cm.10032] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The spindle checkpoint, which prevents anaphase onset upon spindle damage or incorrect chromosome alignment, presents a problem for experimental analysis of protein function in anaphase and cytokinesis. This is because the functional disruption of many proteins before anaphase onset can activate this checkpoint, preventing anaphase and subsequent cell cycle events. This paper compares new and old methods of overriding the spindle checkpoint in prometaphase mammalian tissue culture cells.
Collapse
Affiliation(s)
- Julie C Canman
- University of North Carolina, Department of Biology, 607 Fordham Hall, CB# 3280, Chapel Hill, NC 27599-3280, USA.
| | | | | |
Collapse
|
549
|
Sironi L, Mapelli M, Knapp S, Antoni AD, Jeang KT, Musacchio A. Crystal structure of the tetrameric Mad1-Mad2 core complex: implications of a 'safety belt' binding mechanism for the spindle checkpoint. EMBO J 2002; 21:2496-506. [PMID: 12006501 PMCID: PMC126000 DOI: 10.1093/emboj/21.10.2496] [Citation(s) in RCA: 251] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The spindle checkpoint protein Mad1 recruits Mad2 to unattached kinetochores and is essential for Mad2-Cdc20 complex formation in vivo but not in vitro. The crystal structure of the Mad1-Mad2 complex reveals an asymmetric tetramer, with elongated Mad1 monomers parting from a coiled-coil to form two connected sub-complexes with Mad2. The Mad2 C-terminal tails are hinged mobile elements wrapping around the elongated ligands like molecular 'safety belts'. We show that Mad1 is a competitive inhibitor of the Mad2-Cdc20 complex, and propose that the Mad1-Mad2 complex acts as a regulated gate to control Mad2 release for Cdc20 binding. Mad1-Mad2 is strongly stabilized in the tetramer, but a 1:1 Mad1-Mad2 complex slowly releases Mad2 for Cdc20 binding, driven by favourable binding energies. Thus, the rate of Mad2 binding to Cdc20 during checkpoint activation may be regulated by conformational changes that destabilize the tetrameric Mad1-Mad2 assembly to promote Mad2 release. We also show that unlocking the Mad2 C-terminal tail is required for ligand release from Mad2, and that the 'safety belt' mechanism may prolong the lifetime of Mad2-ligand complexes.
Collapse
Affiliation(s)
| | | | - Stefan Knapp
- Structural Biology Unit, Department of Experimental Oncology, European Institute of Oncology, Via Ripamonti 435, 20141 Milan,
Pharmacia Corporation, Department of Chemistry, Discovery Research Oncology, 20014 Nerviano, Italy and Molecular Virology Section, National Institutes of Health, Bethesda, MD 20892-0460, USA Corresponding author e-mail:
| | | | - Kuan-Teh Jeang
- Structural Biology Unit, Department of Experimental Oncology, European Institute of Oncology, Via Ripamonti 435, 20141 Milan,
Pharmacia Corporation, Department of Chemistry, Discovery Research Oncology, 20014 Nerviano, Italy and Molecular Virology Section, National Institutes of Health, Bethesda, MD 20892-0460, USA Corresponding author e-mail:
| | - Andrea Musacchio
- Structural Biology Unit, Department of Experimental Oncology, European Institute of Oncology, Via Ripamonti 435, 20141 Milan,
Pharmacia Corporation, Department of Chemistry, Discovery Research Oncology, 20014 Nerviano, Italy and Molecular Virology Section, National Institutes of Health, Bethesda, MD 20892-0460, USA Corresponding author e-mail:
| |
Collapse
|
550
|
Saeki A, Tamura S, Ito N, Kiso S, Matsuda Y, Yabuuchi I, Kawata S, Matsuzawa Y. Frequent impairment of the spindle assembly checkpoint in hepatocellular carcinoma. Cancer 2002; 94:2047-54. [PMID: 11932908 DOI: 10.1002/cncr.10448] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Chromosomal instability (CI) leading to aneuploidy is one form of genetic instability, a characteristic feature of various types of cancers. Recent work has suggested that CI can be induced by a spindle assembly checkpoint defect. The aim of the current study was to determine the frequency of a defect of the checkpoint in hepatocellular carcinoma (HCC) and to establish whether alterations of genes encoding the checkpoint were associated with CI in HCC. METHODS Aneuploidy and the function of the spindle assembly checkpoint were examined using DNA flow cytometry and morphologic analysis with microtubule disrupting drugs. To explore the molecular basis, the authors examined the expression and alterations of the mitotic checkpoint gene, BUB1, using Northern hybridization and direct sequencing in 8 HCC cell lines and 50 HCC specimens. Furthermore, the authors examined the alterations of other mitotic checkpoint genes, BUBR1, BUB3, MAD2B, and CDC20, using direct sequencing in HCC cell lines with aneuploidy. RESULTS An impaired spindle assembly checkpoint was found in five (62.5%) of the eight aneuploid cell lines. Transcriptional expressions of the BUB1 gene appeared in all cell lines. While some polymorphic base changes were noted in BUB1, BUBR1, and CDC20, no mutations responsible for impairment of the mitotic checkpoint were found in either the HCC cell lines or HCC specimens, which suggests that these genes did not seem to be involved in tumor development in HCC. CONCLUSIONS The loss of spindle assembly checkpoint occurred with a high frequency in HCC with CI. However, other mechanisms might also contribute to CI in HCC.
Collapse
Affiliation(s)
- Ayuko Saeki
- Department of Internal Medicine and Molecular Science, Graduate School of Medicine, Osaka University, 2-2 B-5 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|