Maeck 2000

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 5

British Journal of Haematology, 2000, 109, 842±846

SHORT REPORT

Genetic instability in myelodysplastic syndrome: detection of


microsatellite instability and loss of heterozygosity in bone
marrow samples with karyotype alterations

Lienhard Maeck, 1 Detlef Haase, 1 Claudia Schoch, 2 Wolfgang Hiddemann 2 and Frauke Alves 1 1 Department
of Haematology and Oncology, Georg-August-University, GoÈttingen, Germany, and 2 Department of Internal Medicine III,
University Hospital Grosshadern, Ludwig-Maximilians-University, MuÈnchen, Germany

Received 2 February 2000; accepted for publication 28 February 2000

Summary. Using a polymerase chain reaction (PCR)- coincidence of LOH, MSI and chromosome abnormalities
based approach, we examined the prevalence of loss of was observed (P , 0´025). Moreover, our data suggest that
heterozygosity (LOH) and microsatellite instability (MSI) in LOH represents an initial rather than a secondary genetic
relation to chromosomal imbalances in myelodysplastic event in MDS, promoting genetic instability in a subset of
syndrome (MDS). Two of 26 patients displayed MSI (8%), patients.
one of them at five loci. LOH was detected in six out of 26
cases (23%), predominantly involving markers IRF1 [5q31] Keywords: myelodysplastic syndrome, microsatellite
and WT1 [11p]. Two patients displayed a corresponding instability, loss of heterozygosity, interferon regulatory
chromosomal deletion by conventional cytogenetics. Sup- factor 1, cytogenetics.
porting the mutator phenotype hypothesis, a significant

The initial genetic event of the presumed multistep malignancies as the consequence of a defective DNA MMR
pathogenesis of myelodysplastic syndrome (MDS) is an machinery. MSI has also been observed in sporadic tumours
unknown submicroscopic stem cell defect, which, in a and distinct haematological disorders, suggesting that DNA
subset of cases, is followed by non-clonal karyotype proofreading failure may be a common pathomechanism in
instability (Haase et al, 1992). In approximately half of the neoplastic transformation.
patients, clonal karyotypic abnormalities, which are detect- To verify whether both mechanisms might be important
able at the stem cell level, occur (Haase et al, 1997). By at the onset of de novo MDS, bone marrow samples were
unravelling loss of heterozygosity (LOH) and microsatellite analysed for LOH and MSI using a panel of 18 microsatellite
instability (MSI), microsatellite polymorphic markers are repeats located on 15 different chromosomal arms. From
useful to test the status of DNA mismatch repair (MMR) and each patient, buccal smears were taken as a source for
cell cycle-conducting tumour-suppressor genes, both of constitutional DNA. The results of the microsatellite
which, if deficient, promote genetic instability (Loeb, 1998). analyses were related to corresponding cytogenetic data.
LOH describes the homozygous state of a distinct
chromosomal region and points to the presence of a closely
located inactivated tumour-suppressor gene that might be PATIENTS AND METHODS
involved in malignant transformation. MSI is defined by the Patients. Twenty three patients with de novo MDS, one
occurrence of novel microsatellite alleles in neoplastic DNA patient with therapy-related MDS (t-MDS) and two patients
when compared with DNA from non-malignant tissue of the with acute myeloid leukaemia secondary to MDS (sAML)
same individual. It is a hallmark of patients with hereditary secondary to MDS (sAML) were recruited for microsatellite
non-polyposis colorectal cancer (HNPCC) and related analyses (Table IA). Bone marrow samples and buccal
smears were collected after informed consent.
Correspondence: F. Alves, Department of Haematology and Cytogenetics. Chromosome analyses were performed using
Oncology, Georg-August-University, Robert-Koch-Str. 40, 37075 a modified GAG-banding technique after short-term cultures
GoÈttingen, Germany. E-mail: falves@gwdg.de of bone marrow samples as described previously (Stollmann

842 q 2000 Blackwell Science Ltd


Table IA. Karyotypes of the patients examined and results of the microsatellite analyses.
q 2000 Blackwell Science Ltd, British Journal of Haematology 109: 842±846

Patient no. Sex Age (years) FAB classification Cytogenetics LOH/MSI

1 M 62 RAEB 46,XY [22]


2 M 57 RAEB 46,XY,del(1)(p33),i(14)(q10) [18]/46,XY [2]
3 F 73 RA 47,XX, 18 [2]/47,XX, 1 11 [1]/46,XX [42]
4 M 61 RAEB 44,XY,dic(1; 22)(p36; p11)(dup(1)(q11p35),del(5)(q13q33),-7,add(17)(p1?1) [9]/
44,XY,dic(1; 22)(p36; p11)(dup(1)(q11p35),del(1)(q11),del(5)(q13q33),-7,add
(17)(p1?1) [3]/46,XY [15]
5 M 62 RAEB 46,XY [25]
6 F 46 t±RAEB 45,XX, 27 [18]/45,XX, 23, 27, 18 [2]/46,XX, 27, 121 [4]/90,XXXX, 27, 27 [1] CACNL1A3, WT1
sporadic chromosomal alterations [12/25]
7 M 72 RAEB 46,XY [1]/45,X,-Y [18]/44,X,-Y,-22 [4] LPL, D10S197,
D12S89, RB1, DCC
8 M 67 RARS 47,XY,t(9; 21)(q13; q22), 1der(9)t(9; 21)(q13; q22) [24]/46,XY [1]
sporadic chromosomal alterations in 5 metaphases
9 F 60 RA 47±50,XX,del(5)(q13),der(?)ins(?; 5)(?; q?) 2,-13, 1der(13)(?), 1del(14)(q13), IRF1, TP53
11 2 3mar [cp24]/additional sporadic chromosomal alterations in 10 metaphases/46,XX [1]
10 M 75 RAEB 46,XY [4]/47,XY, 18 [8]
11 M 64 RAEB-T 46,XY [23]
12 F 62 RAEB 46,XX [10]/44,XX,del(5)(q15q31),-7,-20 [2]/44,XX,del(5)(q15q31),-7,del(17)(p11.2),-20 [2]/
44,XX,del(5)(q13),-7,-20 [4]/44,XX,del(5)(q13),-7,dic(14; 17)(p11; p11), 117,-20 [1]/
80±90,XXXX,idemx2 [cp3]/46,XX,dic(14; 15)(p11; p11) [2]
13 M 59 RARS 46,XY [25]
14 F 73 sAML 46±48,XX, 1der(1), 216, 221, 222, 12 23 mar[cp16]/46,XX [4]
15 M 78 RA 46,XY [25]
16 M 48 46,XY [25]/sporadic chromosomal alterations [9/25] IRF1
17 M 59 CMML 46,XY [25]/sporadic chromosomal alterations [5/25]
18 M 65 sAML 47,XY, 18 [19]/46,XY [1] IRF1
19 M 48 CMML 46,XY [25]; sporadic chromosomal alterations [3/25] D12S89
20 M 80 RAEB±T 46,XY [5]/41±56,XY, ±Y,del(2)(q31),del(3)(p21),del(5)(q15),add(6)(q?), 27,der(9)(q?), 210, 111, 112, IRF1
add(12)(p13),t(12; 15)(p13; q11.2),-16,-18,der(20)t(3; 20)(q21; p13), 11±4Mar [cp19]/occurrence of
polyploid metaphases (10%)
21 M 70 RAEB-T 46,XY [29]/46,XY,inv(1)(p36q21) [1]/continued as clonal abnormality at initial diagnosis in 1993 WT1
22 M 61 RAEB 46,XY del(12)(p11.2) [19]/46,XY [1]
23 M 61 RA 46,XY [25]/sporadic chromosomal alterations [3/25]
24 M 66 RA 46,XY [20]
25 F 65 RA 46,XX [18]

Short Report
26 F 56 RARS 46,XX [25]

FAB, French±American±British classification; RA, refractory anaemia; RARS, refractory anaemia with ring sideroblasts; RAEB, refractory anaemia with excess blasts; CMML. chronic
myelomonocytic leukaemia; RAEB-T, refractory anaemia with excess blasts in transformation; t-RAEB, therapy-related refractory anaemia with excess blasts; sAML, acute myeloid leukaemia
secondary to myelodysplastic syndrome.

843
844 Short Report
Table IB. Loci and primers.

Marker Chromosomal location Length (bp) Repeat motif Primer sequences

BAT-40 1p13.1 , 80±100 TTTT.TT. (T7)¼¼¼.TTTT. (T)40 5 0 -ATTAACTTCCTACACCACAAC-3 0


5 0 -GTAGAGCAAGACCACCTTG-3 0
CACNL1A3 1q31/32 141±157 Not reported 5 0 -GCTGAGCTAGCGAGGGGCAGGGT-3 0
5 0 -CCCAGCAAAAACTGAGTGTGGATG-3 0
BAT-26 2p , 80±100 (T)5¼. (A)26 5 0 -TGACTACTTTTGACTTCAGCC-3 0
5 0 -AACCATTCAACATTTTTAACCC-3 0
D4S171 4q33/35 43±161 (AC)20AG(AGAC)5AGA 5 0 -TGGGTAAAGAGTGAGGCTG-3 0
5 0 -GGTCCAGTAAGAGGACAGT-3 0
APC 5q21/22 96±122 (CA)n 5 0 -ACTCACTCTAGTGATAAATCGGG-3 0
5 0 -AGCAGATAAGACAGTATTACTAGTT-3 0
IRF1 5q31 , 243 (CA)n 5 0 -CTGGAATCTCTATGGCAGATAGGTC-3 0
5 0 -GTGCCCAGGTAGGAAGGGGCTTTAC-3 0
D7S522 7q31 217±229 (CA)n 5 0 -GCCAAACTGCCACTTCTC-3 0
5 0 -ACGTGTTATGCCACTCCC-3 0
D8S87 8p12 145±157 Not reported 5 0 -GGGTTGTTGTAAATTAAAAC-3 0
5 0 -TGTCAAATACTTAAGCACAG-3 0
LPL 8p22 106±134 Not reported 5 0 -TAGAGCACACTATCCAGGTGA-3 0
5 0 -CAGTGGGTTATTTGTGGGATA-3 0
D10S197 10qter 161±173 CACCAGA(CA)7.A.A(CA)12(AGAAA)2 5 0 -ACCACTGCACTTCAGGTGAC-3 0
5 0 -GTGATACTGTCCTCAGGTCTCC-3 0
WT1 11p13 40±148 (CA)n 5 0 -AATGAGACTTACTGGGTGAGG-3 0
5 0 -TTACACAGTAATTTCAAGCAACGG-3 0
D12S89 12p13.2 254±288 (CA)n 5 0 -ATTTGAGAGCAGCGTGTTTT-3 0
5 0 -CCATTATGGGGAGTAGGGGT-3 0
RB1 13q14.3 266±306 (CTTT[T])n 5 0 -CTCCTCCCTACTTACTTGT-3 0
5 0 -AATTAACAAGGTGTGGTGGTACACG-3 0
NF1 17q11.2 171±187 (CA)n 5 0 -CAGAGCAAGACCCTGTCT-3 0
5 0 -CTCCTAACATTTATTAACCTTA-3 0
NM23±H1 17q22 94±104 (CA)n 5 0 -TTGACCGGGGTAGAGAACTC-3 0
5 0 -TCTCAGTACTTCCCGTGACC-3 0
TP53 17p13.1 103±135 (A)14GAAAAGAAAAAGAAAAGAA 5 0 -AGGGATACTATTCAGCCCGAGGTG-3 0
A(.)51(CA)24 5 0 -ACTGCCACTCCTTGCCCCATTC-3 0
DCC 18q21 , 215 Not reported 5 0 -GATGACATTTTCCCTCTAGA-3 0
5 0 -TTTAGTGGTTATTGCCTTGAA-3 0
PLCpr 20q12/13.1 150±184 (CA)n 5 0 -AACCAGTCTGCTCTTCCGGTG-3 0
5 0 -CTGCCTTCAACTGATCTCAATGG-3 0

et al, 1985). Chromosomes were classified according to the (Griffais et al, 1991). The forward primer was designed from
1995 International System for Human Cytogenetic Nomen- the IRF1 genomic DNA sequence (exon 8).
clature. The term sporadic chromosomal alterations is used Aliquots of amplified DNA were mixed with 5 ml of
in cases with unequivocal chromosome and/or chromatid formamide dye solution, heated to 968C for 5 min and then
breaks occurring in at least two metaphases, as well as in chilled on ice before being subjected to electrophoresis on a
cases with sporadic deletions, translocations and inversions 10% polyacrylamide standard denaturing sequencing gel
occurring in only one metaphase. (National Diagnostics, Atlanta, GA, USA). The gels were
DNA isolation. Genomic DNA from bone marrow samples silver-stained using standard procedures.
and buccal smears was isolated by ethanol precipitation Genescan analysis. Analysis was performed according to
following proteinase K digestion using the QIAmp Blood Kit the Genescan 500 TAMRA protocol (ABI, Weiterstadt,
(Qiagen, Hilden, Germany). Germany), using the manufacturer's analysis program
Polymerase chain reaction (PCR). Primer sequences and (ABI).
chromosomal locations of the corresponding microsatellites
are given in Table IB. Primer pairs were synthesized using
RESULTS
an Expedite Model 8009 DNA/RNA synthesizer (Perceptive
Biosystems, Wiesbaden, Germany). Amplifying a GT repeat The results of the microsatellite analyses are summarized in
inside the seventh intron of the IRF1 gene, we introduced a Table IA. Six patients showed LOH (23%) and two patients
new primer pair which was designed according to the displayed MSI (8%). Five of them had advanced stage MDS
octamer frequency disparity method as described previously and one patient had sAML. Case 7, classified as refractory

q 2000 Blackwell Science Ltd, British Journal of Haematology 109: 842±846


Short Report 845
MSI. In contrast, we found a significantly positive correla-
tion among the occurrence of LOH, MSI and abnormal
karyotype, including sporadic chromosomal alterations
(exact Fisher test; P , 0´025). A correlation between loci
affected by cytogenetic anomalies and loci involved in LOH
could be observed in patients 9 and 20. Both displayed
chromosomal deletions within the critical region 5q13±
5q31 and showed LOH of IRF1 (5q31) in bone marrow DNA
(Table IA).

DISCUSSION
A disease-inherent genetic instability is supposed to be the
basis for an accumulation of somatic mutations in MDS
(Haase et al, 1992; Jacobs, 1992). In the present study,
analysing 18 microsatellite repeats located on 15 chromo-
somal arms, LOH was detected in bone marrow DNA from
six patients (23%), whereas two further patients displayed
MSI (8%). One of the latter, classified as RAEB (patient 7),
showed MSI at 5 of 17 informative loci (29%). It is therefore
tempting to suggest that a malfunctioning DNA MMR
machinery might have contributed to the pathogenesis of
MDS in this case.
Previous reports on colorectal cancer determined a
negative correlation between the occurrence of MSI and
other genetic events (Ionov et al, 1993) and suggested MSI
of marker BAT-26 to be a reliable indicator of a replication
error phenotype (Hoang et al, 1997). In our study, MSI and
LOH were associated with clonal karyotypic abnormalities
or sporadic chromosomal alterations, respectively, and none
Fig. 1. (A) MSI detected in patient 7 (RAEB). bm, amplified bone
of our patients presented with alterations of marker BAT-26.
marrow DNA; buc, amplified DNA from corresponding buccal
mucosa. The examined loci and their chromosomal locations are
Although DNA MMR deficiency might be involved in a
designated at the bottom of each paired sample. Allelic gain in bone subset of patients with MDS, different pathogenetic path-
marrow DNA is demonstrated by arrows. (B) Four patients ways seem to be responsible for genetic instability in
displaying LOH at the IRF1 locus. bm, bone marrow DNA; buc, colorectal cancer. This might also explain the considerably
corresponding DNA from buccal epithelium. In contrast to patients higher prevalence of unstable markers in HNPCC (, 40%)
16, 18, and 20, patient 9 displayed loss of the shorter allele in comparison with MDS.
(arrows). (C) Electropherogram showing amplification products of LOH was more frequently detected than MSI, mainly
the IRF1 primers in patient 20. (a) A normal heterozygous signal involving loci IRF1 and WT1. After introducing a new primer
amplified from buccal mucosa; (b) the corresponding bone marrow pair amplifying a GT repeat inside the IRF1 gene, LOH was
sample which exhibits loss of the longer allele. The x-axis shows
detected in four patients. Two of the patients displayed a
fragment sizes in base pairs (bp), the y-axis shows peak height
values. The shorter allele is 248 bp and the longer allele 256 bp
corresponding loss of chromosome band 5q31, representing
long. the minimal commonly deleted region (Le Beau et al, 1993).
Our results underline the possible significance of IRF1 gene
loss in MDS, perhaps as one initial step during pathogenesis.
anaemia with excess blasts (RAEB), showed MSI for five They might also reflect a disease-inherent genetic instability
markers (LPL [8p], D10S197 [10qter], D12S89 [12p], RB1 in the 5q31 region, where IRF1 might not be directly involved
[13q] and DCC [18q]; Fig 1A) and patient 19, who exhibited but is located in the vicinity of another presumed tumour-
chronic myelomonocytic leukaemia (CMML), displayed MSI suppressor gene. Using a primer pair amplifying a CA repeat in
for marker D12S89. Four patients showed LOH at locus the 3 0 untranslated region of the WT1 gene, LOH was detected
IRF1, two at locus WT1 and one patient displayed allelic loss in two patients, one with therapy-related refractory anaemia
for TP53. To verify the relatively high prevalence of LOH at with excess blasts (t-RAEB), which transformed into AML
the IRF1 locus (Fig 1B), bone marrow DNA from patient 20 later on, and one with refractory anaemia with excess blasts
was submitted to a Genescan analysis. By means of this in transformation (RAEB-T). Mutations in the WT1 gene have
additional method, loss of the longer allele (256 bp) in the been reported to occur in 15% of cases of AML and were
bone marrow DNA could be confirmed (Fig 1Ca and b). associated with a poor response to chemotherapy (King-
Underwood et al, 1996). Similarly, allelic loss of WT1 might be
Correlation of karyotype and microsatellite analysis predictive for a poor clinical prognosis in MDS. In view of the
Patients with a normal karyotype showed neither LOH nor predominant localization of LOH at only two loci (IRF1 and

q 2000 Blackwell Science Ltd, British Journal of Haematology 109: 842±846


846 Short Report
WT1), we suggest that the allelic losses detected in our evaluated systematically in order to uncover discrete genetic
patients were initial rather than later genetic events. lesions which would escape conventional cytogenetics. This
Only one of our patients displayed allelic loss at the TP53 could be particularly useful in differentiating those MDS on
locus (case 9). Similarly, it has been shown that p53 a molecular level that otherwise appear homogeneous by
mutations are extremely rare in de novo MDS (Sugimoto et al, conventional karyotyping. In these cases, it would be
1993). We therefore conclude that both karyotype and interesting to see whether patients with MSI or LOH differ
molecular genetic instability in de novo MDS are not in their prognosis from those with normal microsatellite
triggered by cell cycle dysregulation due to loss of the p53 allelotypes.
gene function. This is an intriguing fact as the opposite has
been described in therapy-related leukaemia (Ben-Yehuda
ACKNOWLEDGMENTS
et al, 1996).
Our data showed a significantly positive correlation We thank Dr O. Knobloch from SEQLAB Sequence
between the occurrence of LOH and MSI and an abnormal Laboratories (GoÈttingen, Germany) for performing Genescan
karyotype, including sporadic chromosomal alterations analysis and V. Jassal for helpful comments regarding the
(P , 0´025). These data are in line with the mutator manuscript.
phenotype hypothesis, suggesting that LOH and MSI might
reflect early mutative events which affect mutator genes and
REFERENCES
thus induce genetic changes to a larger extent, i.e.
chromosome instability (Loeb, 1998). However, no correla- Ben-Yehuda, D., Krichevsky, S., Caspi, O., Rund, D., Polliack, A.,
tion was found between chromosomal breakpoints defined Abeliovich, D., Zelig, O., Yahalom, V., Paltiel, O., Or, R., Peretz, T.,
by karyotype analysis and our patients displaying LOH and Ben-Neriah, S., Yehuda, O. & Rachmilewitz, E.A. (1996)
MSI. These data reflect what might be expected when Microsatellite instability and p53 mutations in therapy-related
leukemia suggest mutator phenotype. Blood, 88, 4296±4303.
regarding LOH and MSI as a result of DNA MMR failure
Griffais, R., AndreÂ, P.M. & Thibon, M. (1991) K-tuple frequency in
leading to a general increase in mutation rate. In only two the human genome and polymerase chain reaction. Nucleic Acids
patients displaying LOH, both harbouring a partial deletion Research, 19, 3887±3891.
of chromosome 5, a corresponding defect was found by Haase, D., Fonatsch, C. & Freund, M. (1992) Karyotype instability in
cytogenetic studies and microsatellite analysis. It is probable myelodysplastic syndromes ± a specific step in pathogenesis
that the allelic losses in the remaining four cases were on preceding clonal chromosome anomalies. Leukemia and Lym-
the submicroscopic level. phoma, 8, 221±228.
Two patients with an alteration of the short arm of Haase, D., Feuring-Buske, M., SchaÈfer, C., Schoch, C., Troff, C.,
chromosome 17 displayed no corresponding allelic loss of Gahn, B., Hiddemann, W. & WoÈrmann, B. (1997) Cytogenetic
marker TP53 detected by PCR (patients 4 and 12). Likewise, analysis of CD341 subpopulations in AML and MDS characterized
by the expression of CD38 and CD117. Leukemia, 11, 674±679.
four patients with a monosomy 7 detected by conventional
Hoang, J.M., Cottu, P.H., Thuille, B., Salmon, R.J., Thomas, G. &
cytogenetics displayed no analogous LOH for marker
Hamelin, R. (1997) BAT-26, an indicator of the replication error
D7S522 by PCR (patients 4, 6, 12 and 20). These phenotype in colorectal cancers and cell lines. CancerResearch, 57,
discrepancies may originate from the high percentage of 300±303.
cytogenetically normal cells which in patient 12 amounted Ionov, Y., Peinado, M.A., Malkhosyan, S., Shibata, D. & Perucho, M.
to 63%, in patient 4 to 56% and in patient 20 to 21%. The (1993) Ubiquitous somatic mutations in simple repeated
frequency of LOH and MSI might therefore be under- sequences reveal a new mechanism for colonic carcinogenesis.
estimated in those cases. Nature, 363, 558±561.
In summary, LOH was more frequent than MSI (23% vs. Jacobs, A. (1992) Pathogenesis and clinical variations in the
8%). Although a defective DNA MMR might be affected in a myelodysplastic syndromes. Clinical Haematology, 15, 925±951.
King-Underwood, L., Renshaw, J. & Pritchard-Jones, K. (1996)
small subset of patients with MDS, non-involvement of
Mutations in the Wilms' tumor gene WT1 in leukemias. Blood,
marker BAT-26 and the association of further genetic events
87, 2171±2179.
point to a genetic pathway that is different from the Le Beau, M.M., Espinosa, R., Neuman, W.L., Stock, W., Roulston, D.,
situation in HNPCC. The fact that LOH and MSI exclusively Larson, R.A., Keinanen, M. & Westbrook, C.A. (1993) Cytoge-
occurred in those MDS patients harbouring cytogenetic netic and molecular delineation of the smallest commonly deleted
aberrations, including sporadic chromosomal alterations as region of chromosome 5 in malignant myeloid diseases. Proceed-
well as the predominant localization of LOH at only two loci, ings of the National Academy of Sciences of the United States of
may support the mutator phenotype hypothesis. Thus, America, 90, 5484±5488.
given that LOH and MSI occur at the onset of MDS, they Loeb, L.A. (1998) Cancer cells exhibit a mutator phenotype.
might represent an early predictor for chromosomal Advances in Cancer Research, 72, 25±56.
instability. However, as in only two cases a precise Stollmann, B., Fonatsch, C. & Havers, W. (1985) Persistent Epstein-
Barr virus infection associated with monosomy 7 or chromosome
correlation between karyotypic lesions and submicroscopic
3 abnormality in childhood myeloproliferative disorders. British
events such as LOH could be detected, LOH seems not to be Journal of Haematology, 60, 183±196.
predictive for specific chromosomal lesions. With respect to Sugimoto, K., Hirano, N., Toyoshima, H., Chiba, S.H., Mano, H.,
our results regarding IRF1 and WT1, the critical regions Takaku, F., Yazaki, Y. & &. Hirai, H. (1993) Mutations in p53
affected should be subject to further investigations. More- gene in myelodysplastic syndrome (MDS) and MDS-derived
over, a microsatellite panel specific for MDS should be leukemia. Blood, 81, 3022±3026.

q 2000 Blackwell Science Ltd, British Journal of Haematology 109: 842±846

You might also like