Chromosomal Instability in Cancer: Causes and Consequences

David Gisselsson

Department of Clinical Genetics, University Hospital, SE-221 85 LUND, SWEDEN
david.gisselsson@klingen.lu.se

May 2001

Summary

Many human malignant tumours exhibit abnormal chromosomal segregation at cell division. It is believed that these anomalies play a role in tumorigenesis by increasing the rate of chromosome mutations, including deletion and amplification of genes involved in cellular proliferation and/or survival. In vitro experiments have also shown that mitotic instability may be a mechanism for developing resistance to cytotoxic drugs. Abnormal mitotic mechanisms may result in numerical or structural aberrations in the daughter cells. Numerical aberrations can be caused either by the loss of chromosomes at metaphase/anaphase or by multipolar divisions associated with abnormal number or structure of centrosomes. Structural rearrangements have been associated with chromosomal breakage-fusion-bridge (BFB) cycles that can be initiated by telomeric dysfunction, giving rise to unstable dicentric or ring chromosomes. In most tumours exhibiting chromosomal instability, including high-grade malignant pancreatic, ovarian, and head- and neck carcinomas, as well as osteo- and soft tissue sarcomas, the two types of instability occur together. However, in some low-grade mesenchymal and neuroglial tumours, rarely showing numerical changes, BFB events involving telomeric associations and ring chromosomes dominate the mitotic process. At progression towards higher malignancy in these tumours, complex structural and numerical aberrations become more frequent. This may be explained by a process initiated by telomeric dysfunction and anaphase bridging, which, in turn, may give rise to an increased frequency of multinucleated cells through failure of cytokinesis. These cells will contain an abnormal number of centrosomes leading to multipolar mitotic figures at the next cell division. Further understanding of these events may lead to novel strategies for detection and treatment of malignancy.

Introduction

All malignant tumour types have been shown to contain chromosomal aberrations. The pattern of abnormalities varies greatly between malignancies, ranging from simple balanced rearrangements to complex abnormalities affecting both chromosome structure and number ( Mitelman Database of Chromosome Aberration in Cancer 2001 ). In haematological neoplasms, certain abnormalities are often strongly associated with specific diagnostic entities. Typically, these changes are reciprocal translocations such as the t(9;22) in chronic myelogenous leukaemia (Heim and Mitelman, 1995). Similar genetic abnormalities are seen in some solid tumours, e.g. the 11;22 translocation in Ewing sarcomas and the inversion of proximal 10q in papillary thyroid carcinomas (Vecchio and Santoro, 2000). Such subtype-specific chromosomal abnormalities often correspond to distinct molecular genetic alterations, including the formation of fusion gene products, or the swapping of promoter elements leading to dysregulated gene expression (Åman, 1999). The majority of malignant solid tumours, however, exhibit a complex pattern of chromosomal abnormalities, rarely showing any direct association with specific morphological or prognostic subgroups. Many common aggressive epithelial tumours, such as high-grade pancreatic, ovarian, and lung cancer, fall within this category (Pejovic et al., 1992; Johansson et al., 1995; Gorunova et al., 1998). So do many sarcomas, such as osteosarcoma, leiomysorcoma, and malignant peripheral nerve sheath tumour (Mandahl, 1996). The molecular genetic alterations corresponding to these complex cytogenetic anomalies are not well characterised, although abnormal activation of oncogenes and losses at tumour suppressor genes are common. These changes are rarely subtype specific. However, as a rule, the total number of chromosomal aberrations is roughly proportional to the risk of metastasis (Mitelman et al., 1997).

What is chromosomal instability?

The large number of chromosomal changes frequently seen in malignant solid tumours has brought forth the suggestion that an abnormally high rate of chromosome mutations occurs in neoplastic cells (Nowell, 1976). Tumours exhibiting a large number of chromosomal abnormalities usually also show considerable intratumour variability in the pattern of genetic rearrangements. In most cases, a subset of these abnormalities are shared by all cells of a tumour, indicating that a step-wise accumulation of cytogenetic changes have occurred during tumour growth (Heim et al., 1988). Chromosomal instability may be defined as a state of continuous formation of novel chromosome mutations, at a rate higher than in normal cells. Strictly, this implies that neither cytogenetic complexity, nor cytogenetic heterogeneity per se, should be used as evidence of chromosomal instability, as these states may both be attained through a single mutagenic insult. In practice, instability may be assessed by following the evolution of cytogenetic abnormalities in a tumour cell population over time and by comparing the rate of chromosome mutations with that in a normal cell population (Lengauer et al., 1997). Alternatively, the mutation events can be monitored directly by quantifying the incidence of chromosomal losses, gains, and structural rearrangements at cell division. However, none of these strategies is sufficient to fully evaluate the mechanisms of chromosomal instability. Normally, cells that have undergone DNA breakage are prevented from further proliferation by a number of cell cycle check points. A continuous accumulation of chromosome changes in a cell population may thus arise either through an elevated mutation frequency, a decreased tendency of self-elimination among mutated cells, or both. The scenario is further complicated by the influence of environmental factors on experimental systems (Johansson and Mertens, 1986).

Mechanisms of chromosomal instability

Structural chromosome instability

An elevated frequency of structural chromosome aberrations could be directly caused by an abnormally high incidence of DNA double-strand breaks. Chromosomal breakage can result in a number of different structural rearrangements, some of which give rise to abnormalities of chromosomal segregation at mitosis. For example, terminal deletions due to a break of a single chromatid will result in a centric derivate chromosome plus an acentric fragment. Because of its failure to bind the mitotic spindle, the fragment may be permanently lost in the subsequent cell division, and may be seen as a lagging chromatin body at metaphase or anaphase. Such lagging is a common finding in cell populations exposed to ionising radiation (Natarajan et al., 1996). It has also been described in a number of solid tumours, such as head and neck, and breast carcinomas (Hansemann, 1891; Steinbeck, 1997).

In normal cells, DNA lesions are detected and repaired by a sophisticated physiological machinery. An essential component of this is the BRCA1-associated genome surveillance complex (BASC), including the BRCA1, BRCA2, MSH2, MSH6, MLH1, ATM, BLM proteins, as well as the RAD50 - MRE11 - NBS1 complex (Wang et al. 2000). So far, germ-line mutations of BRCA1, BRCA2, MSH2, MLH1, ATM, and BLM have been found to cause inherited syndromes with an increased tumour incidence (Meyn, 1997). Also, somatic mutations of these genes occur in sporadic tumours, e.g. MSH2 and MLH1 in colorectal carcinomas (Gafa et al., 2000), ATM in lymphomas (Meyn, 1997), and BRCA1 in mammary cancer (Meje, 1995). Both the Bloom syndrome and ataxia telangiectasia exhibit an increased level of spontaneous chromosomal aberrations in somatic cells. Still, the incidence of chromosomal instability far exceeds the frequencies of somatic ATM and BLM mutations in sporadic cancers, indicating that other mechanisms are primarily responsible.

Genomic integrity is also under surveillance from a system of cell cycle checkpoints, preventing cells that have sustained DNA damage from proliferating further. For instance, double-strand DNA breaks induced by clastogenic agents, such as radiation or reactive oxygen species, may lead either to cell cycle arrest or apoptosis by a mechanism including activation of the TP53 and p21 proteins ( Kastan et al., 1991; Bunz et al., 1998; Rotman and Shiloh, 1998; Morrison et al., 2000). Factors involved in this cell cycle checkpoint system are frequently deregulated or inactivated in tumour cells. Mutations of TP53 are responsible for the Li-Fraumeni tumour syndrome and also occur at a frequency of approximately 50% in sporadic malignant tumours. The gene for the TP53-inhibitor MDM2 is amplified in another 10%-20% of bone and soft tissue tumours (Toguchida et al., 1992; Wang et al., 1995; Miller et al., 1996; Lonardo et al., 1997; Yoo et al., 1997). Inactivation of TP53 in immortalised cells results in a marked instability of chromosome structure, including translocations, deletions, telomeric associations, and ring chromosomes (Agapova et al., 1996; Meyn, 1997). A similar pattern of aberrations is typically seen in pancreatic carcinomas and poorly differentiated sarcomas with mutations in TP53 or genomic amplification of MDM2 (Gisselsson et al., 1998; Gisselsson et al., 2000).

The BFB cycle — a chain reaction of chromosomal breakage

Concurrent breaks in two different chromosomes may either give rise to translocations or dicentrics. Whereas translocation derivatives are stably transmitted through cell division, the dicentric chromosomes may be stretched out between the spindle poles to form bridges at anaphase (Figure 1A). These bridges may subsequently break, and the chromosomes are transmitted to the daughter cells with broken ends that may recombine further during the subsequent interphase (McClintock, 1940). Similarly, ring chromosomes having undergone sister chromatid exchange, may be stretched out at anaphase, break, and then be transmitted to the daughter cells as broken chromosomes (Figure 1B; McClintock, 1938). The broken chromosome ends may fuse into novel dicentrics and rings, which may again break at the next cell division. Thus, chromosomal damage may not only result in static aberrations, such as translocations, inversions, deletions, and duplications; it may also result in mitotically unstable chromosomes, which may trigger a series of breakage-fusion-bridge (BFB) events. Such BFB cycles have been shown to occur in many malignant solid tumours with complex chromosome abnormalities, including head and neck, pancreatic, and ovarian carcinomas, as well as leiomyosarcoma, osteosarcoma, malignant fibrous histiocytoma, and atypical lipomatous tumours (Gisselsson et al., 2000; Saunders et al., 2000). Data from in vitro and animal studies indicate that the BFB cycles may be initiated by shortening of telomeric repeat sequences, leading to impaired integrity of chromosome termini and telomeric associations between chromosomes (Artandi and DePinho, 2000). It is probable that similar mechanisms are responsible for the high frequency of BFB instability in human malignancies (Gisselsson et al., unpublished data).

Figure 1 Chromosome breakage-fusion-bridge (BFB) cycles: Dicentric (A) and ring (B) chromosomes may form bridges at anaphase and the broken ends of the two chromatids (red and white) may fuse into novel dicentric and ring-shaped structures in the daughter cells.

Numerical instability

Already at the end of the 19th century, Hansemann observed aberrations of the mitotic apparatus in malignant tumours, including abnormal mitotic polarity and an unequal segregation of chromosomes at anaphase (Hansemann, 1891). A few decades later, Boveri suggested that abnormalities in mitotic polarity could be caused by an abnormal number of centrosomes — the organelles responsible for organisation of the mitotic spindle (Boveri, 1914). Today it is well established that many epithelial tumours exhibit an asymmetrical segregation of chromosomes at the metaphase-anaphase transition, resulting in an aberrant distribution of the genetic material to the daughter cells (Steinbeck, 1998). Also, abnormalities in the number and structure of centrosomes have been observed in malignancies with aneuploid chromosome numbers, including cancers of the breast (Lingle and Salisbury, 1999), colon (Ghadimi et al., 2000), and the head and neck region (Saunders et al., 2000). These changes in centrosomal configuration have been correlated with a number of molecular genetic abnormalities, including amplification of STK15 (Zhou et al., 1998), mutations in TP53 (Carroll et al., 1999), and inactivation of BRCA1 (Xu et al., 1999), BRCA2 (Tutt et al., 1999), and GADD45 (Hollander et al., 1999). Abnormal centrosomal function may also be induced in vitro by expression of the papilloma virus genes E6 and E7, inhibiting normal TP53 and RB1 activity, respectively (Duensing et al., 2000). This is particularly interesting since multipolar mitoses and other manifestations of chromosomal instability are hallmarks of malignant progression in HPV-related cervical carcinomas (Duensing and Münger, 2001). It has also been suggested that inactivation of genes that control the timing of mitotic chromosome segregation may contribute to numerical instability. However, only rare examples of such aberrations have so far been identified (Cahill et al., 1998).

Sine qua non or side effect?

Although chromosomal instability is a common finding in malignant tumours, its precise pathogenetic role remains to be established. Rapid cellular proliferation would undoubtedly be of selective value in a developing tumour. It is likely that cells could increase their mitotic frequency by decreasing the cell cycle intervals spent on DNA repair. An impaired control of cell cycle progression, such as disrupted TP53 or RB1 function, could thereby be coupled to a higher rate of proliferation. These proteins are also involved in the control of genomic integrity and the uncoupling of mitosis from genomic integrity could then merely be a side effect of selection for cells with a high mitotic rate. In this case, a continuous rearrangement of the chromosome complement may even impose a burden on the cell population, and efficient growth would be dependent on a delicate balance between proliferation rate and mutation rate. Functions that stabilise the chromosome complement, while still allowing a permanent disruption of cellular check points, would then be highly favourable for efficient tumour development. It has been suggested that abnormal activation of telomerase could stabilise inherently unstable genomes by synthesising novel telomeric repeats at broken chromosome ends (Artandi and DePinho, 2000). Indeed, telomerase activity has been detected in the majority of malignant epithelial tumours (Krupp et al., 2000).

On the other hand, genomic instability may actually have a selective value per se. An increased rate of chromosomal mutations will result in an elevated frequency of other genetic abnormalities, some if which may directly promote tumour cell proliferation and viability. Such an elevated frequency of chromosomal mutations may even be a prerequisite for oncogenetic processes requiring multiple genetic steps, such as the inactivation of tumour suppressor genes and amplification of oncogenes (Lengauer et al., 1998). Many of the tumours showing genomic instability typically have loss of heterozyogosity for many loci. Also, in tumours showing genomic amplification, BFB events occur at a high frequency. For instance, in head and neck cancer, amplification of the CCND1 gene, located in 11q13, is associated with anaphase bridging of chromosome 11 material (Shuster et al., 2000). These tumours also show loss of heterozyogsity at various tumour suppresser loci, including FHIT in 3p14, INK4A in 9p21, and TP53 in 17p13 (Scully et al., 2000). In atypical lipomatous tumours carrying amplified sequences from 12q13-15, including the SAS, CDK4, HMGIC, and MDM2 genes, the majority of anaphase bridges contain sequences from this particular chromosome segment (Gisselsson et al., 1999). Furthermore, a number of in vitro systems have shown that amplification of genes conferring resistance to cytotoxic drugs may occur through BFB events (Smith et al., 1992; Ma et al., 1993; Coquelle et al., 1997). This suggests that a state of chromosomal instability may not only be crucial for tumour development, but may also play a role in resistance to chemotherapy.

Different mechanisms or steps in a single process?

In most tumours exhibiting chromosomal instability, BFB events and centrosomal abnormalities occur together (Saunders et al., 2000; Gisselsson et al., unpublished data). In analogy, most malignant tumours exhibit both structural and numerical chromosome abnormalities (Mitelman Database of Chromosome Aberrations in Cancer 2001). However, in many low-grade mesenchymal and neuroglial tumours, BFB events involving telomeric associations and ring chromosomes are seen at mitosis, in the absence of major numerical changes (Sawyer et al., 1992, 1998, and 2000; Gisselsson et al., 1999). However, at progression of these tumours towards higher malignancy, numerical aberrations as well as highly complex structural aberrations become more frequent (Mitelman Database of Chromosome Aberrations in Cancer 2001; Rosai et al., 1996). This implies a sequence of parallel cytogenetic and molecular steps, where telomeric dysfunction and BFB events occur at an early stage, and numerical instability develops later. First, a continued proliferation of cells with reduced telomere length (beyond the Hayflick limit) requires an inhibition of cell cycle control mechanisms. In vitro, this may be induced by partial inhibition of normal TP53 and RB1 function, for instance by SV40 transfection (Jha et al., 1998). The corresponding in vivo changes remain to be elucidated, although recent findings indicate that some cell types, such as human mammary epithelial cells, may spontaneously proliferate beyond the normal telomere length and acquire chromosomal changes such as dicentrics and rings (Romanov et al., 2001). Further survival of cell populations with disrupted telomeric integrity, resulting in frequent BFB events, would require additional impairment or total abrogation of the systems normally causing arrest or apoptosis in cells with double-strand DNA breaks. This step appears to be associated with inactivating mutations in TP53 or high-level amplification of MDM2 (Gisselsson, 2000). Also, the numerical chromosome instability associated with abnormal centrosome function indicates that highly malignant cells have acquired some tolerance to massive genomic imbalances.

Figure 2
Hypothetical scenario of progressive mitotic instability: Telomere shortening, seen as absence of detectable TTAGGG repeats (top image; green fluorescence) may compromise the integrity of chromosome ends, leading to the formation of rings and dicentrics. These may form bridges at anaphase, which either breaks and initiate a series of BFB-events, or induce cytokinetic failure leading to the formation of binucleate cells with supernumerary centrosomes. Cells with an abnormal centrosome number (orange fluorescence) may form multipolar mitoses at the next cell division. Thus, telomeric dysfunction may result both in structural and numerical chromosome instability.

The common concurrence of BFB instability and centrosome abnormalities suggests that these phenomena are mechanistically linked. Although it is true that both these instabilities may be associated with similar molecular genetic lesions, such as TP53 mutation, their causal relationship, if any, remains unclear. There may be one rather straightforward relationship, however. It is well established that anaphase bridging may cause collapse of the cytokinetic process, leading to formation of cells with a duplicated genome (McClintock, 1938). Tumours with BFB events show a high frequency of binucleated cells (Gisselsson et al., 2001). These cells would not only carry the double amount of genetic material, but also twice the normal number of centrosomes. After the next round of replication, such cells may thus enter mitosis with abnormal centrosome configurations, leading to either tri- or tetrapolar cell divisions (Figure 2). Incomplete cytokinesis could then easily explain the connection between telomere shortening and BFB events, on one hand, and mitotic multipolarity, on the other hand.

Acknowledgements

The author is grateful to Professor Nils Mandahl, Lund University Hospital, Dr. Stefan Duensing, Harvard Medical School, and Dr. Sergey Proshin, Russian Academy of Sciences, for valuable discussions and critical reading of the manuscript.

Bibliography

Chromosome changes caused by alterations of p53 expression.
Agapova LS, Ilyinskaya GV, Turovets NA, Ivanov AV, Chumakov PM, Kopnin BP
Mutation research. 1996 ; 354 (1) : 129-138.
PMID 8692199
 
A critical role for telomeres in suppressing and facilitating carcinogenesis.
Artandi SE, DePinho RA
Current opinion in genetics & development. 2000 ; 10 (1) : 39-46.
PMID 10679392
 
Zur Frage der Entstehung maligner Tumoren. Verlag von Gustav Fischer, Jena
Boveri T
1914.
 
Requirement for p53 and p21 to sustain G2 arrest after DNA damage.
Bunz F, Dutriaux A, Lengauer C, Waldman T, Zhou S, Brown JP, Sedivy JM, Kinzler KW, Vogelstein B
Science (New York, N.Y.). 1998 ; 282 (5393) : 1497-1501.
PMID 9822382
 
Mutations of mitotic checkpoint genes in human cancers.
Cahill DP, Lengauer C, Yu J, Riggins GJ, Willson JK, Markowitz SD, Kinzler KW, Vogelstein B
Nature. 1998 ; 392 (6673) : 300-303.
PMID 9521327
 
Centrosome hyperamplification in human cancer: chromosome instability induced by p53 mutation and/or Mdm2 overexpression.
Carroll PE, Okuda M, Horn HF, Biddinger P, Stambrook PJ, Gleich LL, Li YQ, Tarapore P, Fukasawa K
Oncogene. 1999 ; 18 (11) : 1935-1944.
PMID 10208415
 
Expression of fragile sites triggers intrachromosomal mammalian gene amplification and sets boundaries to early amplicons.
Coquelle A, Pipiras E, Toledo F, Buttin G, Debatisse M
Cell. 1997 ; 89 (2) : 215-225.
PMID 9108477
 
The human papillomavirus type 16 E6 and E7 oncoproteins cooperate to induce mitotic defects and genomic instability by uncoupling centrosome duplication from the cell division cycle.
Duensing S, Lee LY, Duensing A, Basile J, Piboonniyom S, Gonzalez S, Crum CP, Munger K
Proceedings of the National Academy of Sciences of the United States of America. 2000 ; 97 (18) : 10002-10007.
PMID 10944189
 
Centrosome abnormalities, genomic instability and carcinogenic progression.
Duensing S, Münger K
Biochimica et biophysica acta. 2001 ; 1471 (2) : M81-M88.
PMID 11342187
 
Sporadic colorectal adenocarcinomas with high-frequency microsatellite instability.
Gafà R, Maestri I, Matteuzzi M, Santini A, Ferretti S, Cavazzini L, Lanza G
Cancer. 2000 ; 89 (10) : 2025-2037.
PMID 11066042
 
Centrosome amplification and instability occurs exclusively in aneuploid, but not in diploid colorectal cancer cell lines, and correlates with numerical chromosomal aberrations.
Ghadimi BM, Sackett DL, Difilippantonio MJ, Schröck E, Neumann T, Jauho A, Auer G, Ried T
Genes, chromosomes & cancer. 2000 ; 27 (2) : 183-190.
PMID 10612807
 
Chromosomal Instability and Genomic Amplification in Bone and Soft Tissue Tumours. Lund University Doctoral Thesis, Lund
Gisselsson D
2000.
 
Abnormal nuclear shape in solid tumors reflects mitotic instability.
Gisselsson D, Björk J, Höglund M, Mertens F, Dal Cin P, Akerman M, Mandahl N
The American journal of pathology. 2001 ; 158 (1) : 199-206.
PMID 11141493
 
The structure and dynamics of ring chromosomes in human neoplastic and non-neoplastic cells.
Gisselsson D, Höglund M, Mertens F, Johansson B, Dal Cin P, Van den Berghe H, Earnshaw WC, Mitelman F, Mandahl N
Human genetics. 1999 ; 104 (4) : 315-325.
PMID 10369161
 
Chromosomal organization of amplified chromosome 12 sequences in mesenchymal tumors detected by fluorescence in situ hybridization.
Gisselsson D, Höglund M, Mertens F, Mitelman F, Mandahl N
Genes, chromosomes & cancer. 1998 ; 23 (3) : 203-212.
PMID 9790500
 
Chromosomal breakage-fusion-bridge events cause genetic intratumor heterogeneity.
Gisselsson D, Pettersson L, Höglund M, Heidenblad M, Gorunova L, Wiegant J, Mertens F, Dal Cin P, Mitelman F, Mandahl N
Proceedings of the National Academy of Sciences of the United States of America. 2000 ; 97 (10) : 5357-5362.
PMID 10805796
 
Cytogenetic analysis of pancreatic carcinomas: intratumor heterogeneity and nonrandom pattern of chromosome aberrations.
Gorunova L, Höglund M, Andrén-Sandberg A, Dawiskiba S, Jin Y, Mitelman F, Johansson B
Genes, chromosomes & cancer. 1998 ; 23 (2) : 81-99.
PMID 9739011
 
Ueber patologische Mitosen
Hansemann D
Arch Pathol Anat Phys Klin Med. 18 ; 119 : 299-326.
 
Genetic convergence and divergence in tumor progression.
Heim S, Mandahl N, Mitelman F
Cancer research. 1988 ; 48 (21) : 5911-5916.
PMID 3048652
 
Cancer Cytogenetics
Heim S, Mitelman F
2nd Ed. Wiley-Liss, New York. 1995.
 
Genomic instability in Gadd45a-deficient mice.
Hollander MC, Sheikh MS, Bulavin DV, Lundgren K, Augeri-Henmueller L, Shehee R, Molinaro TA, Kim KE, Tolosa E, Ashwell JD, Rosenberg MP, Zhan Q, Fern´ndez-Salguero PM, Morgan WF, Deng CX, Fornace AJ Jr
Nature genetics. 1999 ; 23 (2) : 176-184.
PMID 10508513
 
SV40-Mediated immortalization.
Jha KK, Banga S, Palejwala V, Ozer HL
Experimental cell research. 1998 ; 245 (1) : 1-7.
PMID 9828095
 
Influence of serum concentration on chromosome breakage frequency.
Johansson B, Mertens F
Hereditas. 1986 ; 104 (1) : 111-112.
PMID 3710823
 
Cytogenetic analysis of short-term cultured squamous cell carcinomas of the lung.
Johansson M, Jin Y, Mandahl N, Hambraeus G, Johansson L, Mitelman F, Heim S
Cancer genetics and cytogenetics. 1995 ; 81 (1) : 46-55.
PMID 7773960
 
Participation of p53 protein in the cellular response to DNA damage.
Kastan MB, Onyekwere O, Sidransky D, Vogelstein B, Craig RW
Cancer research. 1991 ; 51 (23 Pt 1) : 6304-6311.
PMID 1933891
 
Telomerase, immortality and cancer.
Krupp G, Bonatz G, Parwaresch R
Biotechnology annual review. 2000 ; 6 : 103-140.
PMID 11193292
 
Genetic instability in colorectal cancers.
Lengauer C, Kinzler KW, Vogelstein B
Nature. 1997 ; 386 (6625) : 623-627.
PMID 9121588
 
Genetic instabilities in human cancers.
Lengauer C, Kinzler KW, Vogelstein B
Nature. 1998 ; 396 (6712) : 643-649.
PMID 9872311
 
Altered centrosome structure is associated with abnormal mitoses in human breast tumors.
Lingle WL, Salisbury JL
The American journal of pathology. 1999 ; 155 (6) : 1941-1951.
PMID 10595924
 
p53 and MDM2 alterations in osteosarcomas: correlation with clinicopathologic features and proliferative rate.
Lonardo F, Ueda T, Huvos AG, Healey J, Ladanyi M
Cancer. 1997 ; 79 (8) : 1541-1547.
PMID 9118036
 
Sister chromatid fusion initiates amplification of the dihydrofolate reductase gene in Chinese hamster cells.
Ma C, Martin S, Trask B, Hamlin JL
Genes & development. 1993 ; 7 (4) : 605-620.
PMID 8458577
 
Cytogenetics and molecular genetics of bone and soft tissue tumors.
Mandahl N
Advances in cancer research. 1996 ; 69 : 63-99.
PMID 8791679
 
The Production of Homozygous Deficient Tissues with Mutant Characteristics by Means of the Aberrant Mitotic Behavior of Ring-Shaped Chromosomes.
McClintock B
Genetics. 1938 ; 23 (4) : 315-376.
PMID 17246891
 
The Stability of Broken Ends of Chromosomes in Zea Mays.
McClintock B
Genetics. 1941 ; 26 (2) : 234-282.
PMID 17247004
 
Chromosome instability syndromes: lessons for carcinogenesis.
Meyn MS
Current topics in microbiology and immunology. 1997 ; 221 : 71-148.
PMID 8979441
 
Alterations of the p53, Rb and MDM2 genes in osteosarcoma.
Miller CW, Aslo A, Won A, Tan M, Lampkin B, Koeffler HP
Journal of cancer research and clinical oncology. 1996 ; 122 (9) : 559-565.
PMID 8781571
 
Clinical significance of cytogenetic findings in solid tumors.
Mitelman F, Johansson B, Mandahl N, Mertens F
Cancer genetics and cytogenetics. 1997 ; 95 (1) : 1-8.
PMID 9140447
 
Mitelman Database of Chromosome Aberrations in Cancer
http://cgapncinihgov/Chromosomes/Mitelman
 
The controlling role of ATM in homologous recombinational repair of DNA damage.
Morrison C, Sonoda E, Takao N, Shinohara A, Yamamoto K, Takeda S
The EMBO journal. 2000 ; 19 (3) : 463-471.
PMID 10654944
 
Current cytogenetic methods for detecting exposure and effects of mutagens and carcinogens.
Natarajan AT, Boei JJ, Darroudi F, Van Diemen PC, Dulout F, Hande MP, Ramalho AT
Environmental health perspectives. 1996 ; 104 Suppl 3 : 445-448.
PMID 8781361
 
The clonal evolution of tumor cell populations.
Nowell PC
Science (New York, N.Y.). 1976 ; 194 (4260) : 23-28.
PMID 959840
 
Chromosome aberrations in 35 primary ovarian carcinomas.
Pejovic T, Heim S, Mandahl N, Baldetorp B, Elmfors B, Flodérus UM, Furgyik S, Helm G, Himmelmann A, Willén H
Genes, chromosomes & cancer. 1992 ; 4 (1) : 58-68.
PMID 1377010
 
Normal human mammary epithelial cells spontaneously escape senescence and acquire genomic changes.
Romanov SR, Kozakiewicz BK, Holst CR, Stampfer MR, Haupt LM, Tlsty TD
Nature. 2001 ; 409 (6820) : 633-637.
PMID 11214324
 
Combined morphologic and karyotypic study of 59 atypical lipomatous tumors. Evaluation of their relationship and differential diagnosis with other adipose tissue tumors (a report of the CHAMP Study Group).
Rosai J, Akerman M, Dal Cin P, DeWever I, Fletcher CD, Mandahl N, Mertens F, Mitelman F, Rydholm A, Sciot R, Tallini G, Van den Berghe H, Van de Ven W, Vanni R, Willen H
The American journal of surgical pathology. 1996 ; 20 (10) : 1182-1189.
PMID 8827023
 
ATM: from gene to function.
Rotman G, Shiloh Y
Human molecular genetics. 1998 ; 7 (10) : 1555-1563.
PMID 9735376
 
Chromosomal instability and cytoskeletal defects in oral cancer cells.
Saunders WS, Shuster M, Huang X, Gharaibeh B, Enyenihi AH, Petersen I, Gollin SM
Proceedings of the National Academy of Sciences of the United States of America. 2000 ; 97 (1) : 303-308.
PMID 10618413
 
A role for telomeric and centromeric instability in the progression of chromosome aberrations in meningioma patients.
Sawyer JR, Husain M, Pravdenkova S, Krisht A, Al-Mefty O
Cancer. 2000 ; 88 (2) : 440-453.
PMID 10640979
 
Clonal telomeric fusions and chromosome instability in a subcutaneous sacrococcygeal myxopapillary ependymoma.
Sawyer JR, Miller JP, Ellison DA
Cancer genetics and cytogenetics. 1998 ; 100 (2) : 169-175.
PMID 9428364
 
Telomeric associations evolving to ring chromosomes in a recurrent pleomorphic xanthoastrocytoma.
Sawyer JR, Thomas EL, Roloson GJ, Chadduck WM, Boop FA
Cancer genetics and cytogenetics. 1992 ; 60 (2) : 152-157.
PMID 1606558
 
Genetic aberrations in oral or head and neck squamous cell carcinoma 3: clinico-pathological applications.
Scully C, Field JK, Tanzawa H
Oral oncology. 2000 ; 36 (5) : 404-413.
PMID 10964046
 
A consistent pattern of RIN1 rearrangements in oral squamous cell carcinoma cell lines supports a breakage-fusion-bridge cycle model for 11q13 amplification.
Shuster MI, Han L, Le Beau MM, Davis E, Sawicki M, Lese CM, Park NH, Colicelli J, Gollin SM
Genes, chromosomes & cancer. 2000 ; 28 (2) : 153-163.
PMID 10825000
 
Fusions near telomeres occur very early in the amplification of CAD genes in Syrian hamster cells.
Smith KA, Stark MB, Gorman PA, Stark GR
Proceedings of the National Academy of Sciences of the United States of America. 1992 ; 89 (12) : 5427-5431.
PMID 1351682
 
Imbalance of morphologically addressed telophases reflects interphase DNA aneuploidy in tumorigenesis.
Steinbeck RG
European journal of histochemistry : EJH. 1997 ; 41 (4) : 243-254.
PMID 9491310
 
Chromosome division figures reveal genomic instability in tumorigenesis of human colon mucosa.
Steinbeck RG
British journal of cancer. 1998 ; 77 (7) : 1027-1033.
PMID 9569034
 
Mutation spectrum of the p53 gene in bone and soft tissue sarcomas.
Toguchida J, Yamaguchi T, Ritchie B, Beauchamp RL, Dayton SH, Herrera GE, Yamamuro T, Kotoura Y, Sasaki MS, Little JB
Cancer research. 1992 ; 52 (22) : 6194-6199.
PMID 1423262
 
Absence of Brca2 causes genome instability by chromosome breakage and loss associated with centrosome amplification.
Tutt A, Gabriel A, Bertwistle D, Connor F, Paterson H, Peacock J, Ross G, Ashworth A
Current biology : CB. 1999 ; 9 (19) : 1107-1110.
PMID 10531007
 
Abnormalities of p53 and p110RB tumor suppressor gene expression in human soft tissue tumors: correlations with cell proliferation and tumor grade.
Wang J, Coltrera MD, Gown AM
Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc. 1995 ; 8 (8) : 837-842.
PMID 8552572
 
BASC, a super complex of BRCA1-associated proteins involved in the recognition and repair of aberrant DNA structures.
Wang Y, Cortez D, Yazdi P, Neff N, Elledge SJ, Qin J
Genes & development. 2000 ; 14 (8) : 927-939.
PMID 10783165
 
Oncogenes and thyroid cancer.
Vecchio G, Santoro M
Clinical chemistry and laboratory medicine : CCLM / FESCC. 2000 ; 38 (2) : 113-116.
PMID 10834397
 
Centrosome amplification and a defective G2-M cell cycle checkpoint induce genetic instability in BRCA1 exon 11 isoform-deficient cells.
Xu X, Weaver Z, Linke SP, Li C, Gotay J, Wang XW, Harris CC, Ried T, Deng CX
Molecular cell. 1999 ; 3 (3) : 389-395.
PMID 10198641
 
p53 gene mutations and p53 protein expression in human soft tissue sarcomas.
Yoo J, Lee HK, Kang CS, Park WS, Lee JY, Shim SI
Archives of pathology & laboratory medicine. 1997 ; 121 (4) : 395-399.
PMID 9140310
 
Tumour amplified kinase STK15/BTAK induces centrosome amplification, aneuploidy and transformation.
Zhou H, Kuang J, Zhong L, Kuo WL, Gray JW, Sahin A, Brinkley BR, Sen S
Nature genetics. 1998 ; 20 (2) : 189-193.
PMID 9771714
 
Fusion genes in solid tumors.
Aman P
Seminars in cancer biology. 1999 ; 9 (4) : 303-318.
PMID 10448117
 
Written2000-02Antonio Cuneo
Section, Dept. Of Biomedical Sciences, University of Ferrara, 44100 Ferrara Italy
Updated2001-05David Gisselsson
of Clinical Genetics, University Hospital, SE-221 85 Lund, Sweden

Citation

This paper should be referenced as such :
Gisselsson D
Chromosomal Instability in Cancer: Causes and Consequence
Atlas Genet Cytogenet Oncol Haematol. in press
On line version : http://AtlasGeneticsOncology.org/Deep/ChromosomInstabilID20023.htm
History of this paper:
Gisselsson, D. Chromosomal instability in cancer: Causes, consequences. Atlas Genet Cytogenet Oncol Haematol. 2001;5(3):236-243.
http://documents.irevues.inist.fr/bitstream/handle/2042/37771/05-2001-ChromosomInstabilID20023.pdf