jumping translocations of 1q in myelodysplastic syndrome and … · 2019. 7. 30. · 9 mds 8 1q12 2...

6
Case Report Jumping Translocations of 1q in Myelodysplastic Syndrome and Acute Myeloid Leukemia: Report of Three Cases and Review of Literature T. Couture, 1,2 K. Amato, 1 A. DiAdamo, 1 and P. Li 1 1 Clinical Cytogenetics Laboratory, Department of Genetics, Yale School of Medicine, New Haven, CT, USA 2 Diagnostic Genetics Sciences Program, Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA Correspondence should be addressed to P. Li; [email protected] Received 2 July 2018; Accepted 15 August 2018; Published 9 September 2018 Academic Editor: Christos Yapijakis Copyright © 2018 T. Couture et al. is is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Jumping translocations of 1q refer to the break-off of chromosome 1q as a donor fusing to two or more recipient chromosomes. We detected jumping translocations of 1q in three patients with initial diagnosis of myelodysplastic syndrome (MDS) and later progression to acute myeloid leukemia (AML). Review of literature found jumping translocations of 1q in 30 reported cases of MDS and AML. e cytogenetic findings from these 33 cases showed that seven cases had a stemline clone and 26 cases had de novo jumping translocations of 1q in which 5% of cell lineages had additional structural rearrangements. In 75% of cases, the 1q donor jumped to the short arm of recipient acrocentric chromosomes. Approximately 82% of the fusions occurred in the telomeric regions of short and long arms and 18% occurred in the pericentric or interstitial regions of recipient chromosomes. Hypomethylation of the donor 1q pericentromeric region and shortened telomeres in recipient chromosomes were associated with the formation of jumping translocations. Jumping translocations of 1q as an indication of chromosomal instability pose high risk for progression of MDS to AML and a poor prognosis. Further understanding of underlying genomic defects and their clinical significance will improve overall treatment and patient care. 1. Introduction Jumping translocations are a rare type of cytogenetic abnor- mality initially detected in a case of acute monocytic leukemia and later found in various types of leukemias [1–3]. Jumping translocations occur when one donor chromosome segment breaks off and fuses to two or more recipient chromosomes in a successive manner and result in clonal abnormalities with a few cell lineages sharing a gain of the donor chromosome segment. e fusions of the break-off donor chromosome segment to telomeric or interstitial regions of recipient chro- mosomes form different chromosomal patterns of jumping translocations from different patients and among different types of leukemias [1–3]. e most commonly seen jumping translocations involving a 1q as the donor chromosome segment are referred to as jumping translocations of 1q. In this report, we present cytogenetic findings of jumping translocations of 1q from three cases initially diagnosed with myelodysplastic syndrome (MDS) and later progressed to acute myeloid leukemia (AML) and the additional 30 cases from review of the literature [1–10]. ese results provide more information of the chromosomal patterns, underlying formation mechanisms, and clinical significance of jumping translocations of 1q in MDS and AML. 2. Case Presentation and Review of the Literature A retrospective review of cases with MDS and AML from the CytoAccess database in Yale Cytogenetics Laboratory found three cases with jumping translocations of 1q [11]. Chromosome and fluorescence in situ hybridization (FISH) analyses were performed following laboratory’s standardized protocols. Review of literature specific for jumping transloca- tions of 1q in MDS and AML found 30 cases from ten reports Hindawi Case Reports in Genetics Volume 2018, Article ID 8296478, 5 pages https://doi.org/10.1155/2018/8296478

Upload: others

Post on 21-Aug-2021

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Jumping Translocations of 1q in Myelodysplastic Syndrome and … · 2019. 7. 30. · 9 MDS 8 1q12 2 10 MDS 8 1q31 2 11 MDS 8 1q12 2 12 MDS 8 1q12 4 13 MDS 8 1q12 2 14 MDS 8 1q21 5

Case ReportJumping Translocations of 1q in MyelodysplasticSyndrome and Acute Myeloid Leukemia: Report of Three Casesand Review of Literature

T. Couture,1,2 K. Amato,1 A. DiAdamo,1 and P. Li 1

1Clinical Cytogenetics Laboratory, Department of Genetics, Yale School of Medicine, New Haven, CT, USA2Diagnostic Genetics Sciences Program, Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA

Correspondence should be addressed to P. Li; [email protected]

Received 2 July 2018; Accepted 15 August 2018; Published 9 September 2018

Academic Editor: Christos Yapijakis

Copyright © 2018 T. Couture et al. This is an open access article distributed under the Creative Commons Attribution License,which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Jumping translocations of 1q refer to the break-off of chromosome 1q as a donor fusing to two or more recipient chromosomes.We detected jumping translocations of 1q in three patients with initial diagnosis of myelodysplastic syndrome (MDS) and laterprogression to acutemyeloid leukemia (AML). Review of literature found jumping translocations of 1q in 30 reported cases of MDSand AML. The cytogenetic findings from these 33 cases showed that seven cases had a stemline clone and 26 cases had de novojumping translocations of 1q in which 5% of cell lineages had additional structural rearrangements. In 75% of cases, the 1q donorjumped to the short arm of recipient acrocentric chromosomes. Approximately 82% of the fusions occurred in the telomeric regionsof short and long arms and 18% occurred in the pericentric or interstitial regions of recipient chromosomes. Hypomethylation ofthe donor 1q pericentromeric region and shortened telomeres in recipient chromosomes were associated with the formation ofjumping translocations. Jumping translocations of 1q as an indication of chromosomal instability pose high risk for progressionof MDS to AML and a poor prognosis. Further understanding of underlying genomic defects and their clinical significance willimprove overall treatment and patient care.

1. Introduction

Jumping translocations are a rare type of cytogenetic abnor-mality initially detected in a case of acutemonocytic leukemiaand later found in various types of leukemias [1–3]. Jumpingtranslocations occur when one donor chromosome segmentbreaks off and fuses to two ormore recipient chromosomes ina successive manner and result in clonal abnormalities witha few cell lineages sharing a gain of the donor chromosomesegment. The fusions of the break-off donor chromosomesegment to telomeric or interstitial regions of recipient chro-mosomes form different chromosomal patterns of jumpingtranslocations from different patients and among differenttypes of leukemias [1–3]. The most commonly seen jumpingtranslocations involving a 1q as the donor chromosomesegment are referred to as jumping translocations of 1q.In this report, we present cytogenetic findings of jumpingtranslocations of 1q from three cases initially diagnosed with

myelodysplastic syndrome (MDS) and later progressed toacute myeloid leukemia (AML) and the additional 30 casesfrom review of the literature [1–10]. These results providemore information of the chromosomal patterns, underlyingformation mechanisms, and clinical significance of jumpingtranslocations of 1q in MDS and AML.

2. Case Presentation and Review ofthe Literature

A retrospective review of cases with MDS and AML fromthe CytoAccess database in Yale Cytogenetics Laboratoryfound three cases with jumping translocations of 1q [11].Chromosome and fluorescence in situ hybridization (FISH)analyses were performed following laboratory’s standardizedprotocols. Review of literature specific for jumping transloca-tions of 1q in MDS and AML found 30 cases from ten reports

HindawiCase Reports in GeneticsVolume 2018, Article ID 8296478, 5 pageshttps://doi.org/10.1155/2018/8296478

Page 2: Jumping Translocations of 1q in Myelodysplastic Syndrome and … · 2019. 7. 30. · 9 MDS 8 1q12 2 10 MDS 8 1q31 2 11 MDS 8 1q12 2 12 MDS 8 1q12 4 13 MDS 8 1q12 2 14 MDS 8 1q21 5

2 Case Reports in Genetics

[1–10]. Two caseswith jumping translocations involving otherdonor chromosomes of 11q and 13q12 [7] and one case withjumping translocations of 3q13.1 were excluded [10].

2.1. Case 1. Thismale patient was first evaluated at age 59withan indication of leukopenia and mild anemia; cytogeneticanalysis found a normal result. Two years later, this patientwas diagnosed with MDS and chromosomal analysis ona bone marrow aspirate showed an abnormal clone withjumping translocations of 1q. The karyotype was as follows:46,XY,der(15)t(1;15)(q12;p12)[7]/46,XY,der(21)t(1;21)(q12;p12)[3]/46,XY,dup(1)(q21q42)[2]/46,XY,der(14)t(1;14)(q12;p12)[2]/46,XY[6]. FISH analysis using dual color probes for thePBX1 gene at 1q23.3 and TCF3 gene at 19p13.3 detectedthree copies of 1q in 32% of bone marrow cells. Thispatient started chemotherapy and then received a bonemarrow transplant in two months. Six months later,the patient progressed to AML and died at age 62.Final chromosomal analysis revealed a karyotype of46,XY,der(1;15)(q12;p12),del(20)(q11.2q13.3)[15]. FISH testsconfirmed three copies of 1q and a deletion at 20q in83.5%-96.5% of bone marrow cells.

2.2. Case 2. This patient, a 63-year old male, was referredwith MDS by an indication of MDS with excess blasts. Chro-mosomal analysis showed a karyotype of 46,XY,der(15)t(1;15)(q12;p11.2)[4]/46,XY,der(22)t(1;22)(q12;p11.2)[2]/46,XY[7].FISH analysis using dual color probes for the CDKN2C geneat 1p32.3 and CKS1B gene at 1q21.3 confirmed three copies of1q in 5.5% of bone marrow cells. Over the next six months,the percentage of abnormal cells increased despite tworounds of chemotherapy and the patient progressed to AML.A transit clone with the der(15)t(1;15) and a deletion at 11q wasnoted. The last chromosome analysis revealed a karyotype of46,XY,der(15)t(1;15)(q12;p11.2)[13]/46,XY,der(22)t(1;22)(q12;p11.2)[2]. FISH detected three copies of 1q in 94% of bonemarrow cells. This patient died eight months after the initialfinding of the jumping translocations of 1q.

2.3. Case 3. This patient was a male at age 73 when diagnosedwith MDS. Chromosomal analysis showed a karyotypeof 46,XY,der(15)t(1;15)(q12;p11.2)[13]/46,XY[7]. FISH con-firmed three copies of 1q in 82.5% of bone marrow cells. Twoyears later, the patient progressed to AML and died soonafter the last chromosome analysis which revealed a karyo-type of 46,XY,der(15)t(1;15)(q12;p11.2)[9]/46,XY,der(22)t(1;22)(q12;p11.2)[4]/46,X,der(Y)t(Y;1)(q12;q11),t(10;17)(p10;p10)[3].FISH detected three copies of 1q in 94% of bone marrowcells.

The cytogenetic findings of successive studies from thesethree patients all showed persistent jumping translocationsof 1q with an increased percentage in bone marrow cells.The chromosome patterns of jumping translocations of 1qare shown in Figure 1(a). Review of literature specific forjumping translocations of 1q in MDS and AML found 30cases from ten reports [1–10]. The clinical indications andpatterns of jumping translocation of 1q from these 33 cases aresummarized in Figure 2. For a total of 110 cell lineages fromthese 33 cases, the cell lineages with jumping translocations

of 1q in a patient ranged from two to ten, with an averageof about three cell lineages. Stemline abnormalities beforethe emerging of jumping translocations of 1q were noted inseven cases (No. 2-7, 30), seemingly unrelated clones werenoted in two cases (No. 8 and 27), related clones with ajumping translocation and an additional rearrangement werenoted in two cases (No. 29 and 33), an evolved abnormalclone with an additional aberration of a deletion in a 20qwas noted in one case (No. 31), and a transit abnormal clonewith an additional aberration of a deletion in an 11q wasnoted in one case (No. 32).These results indicated that at least39% (13/33) of cases with jumping translocations of 1q hadother clonal abnormalities. However, in cases with de novojumping translocations of 1q, other structural rearrangementsare rare events and only occur in 15% (4/26) of cases and inabout 5% (4/79) of cell lineages with jumping translocationsof 1q. The donor chromosome of 1q showed breakpoints inthe pericentric region of 1q11, q12, and q21 except for onecase with the breakpoint at 1q31q32. In terms of recipientchromosomes, 82% of the fusions occurred most likely inthe telomeric regions of short (p) and long (q) arms and18% occurred in the pericentric or interstitial regions of achromosome. The most frequently seen telomeric fusionswere 15p (12%), 22p (11%), 21p (8%), 14p (7%), 13p (4%), Yq(4%), 17q (4%), 18q (4%), and 21q (3%). The most frequentlyseen pericentric or interstitial fusions were 16q11.2q12 (4%),7q11 (3%), and 20q12q13.1 (3%). Jumping translocations of 1qfusing with the short-arm telomeric regions of at least one ofthe acrocentric chromosomes 13, 14, 15, 21, and 22 were notedin 75% (25/33) of cases.

3. Discussion

The results from these 33 cases revealed the chromoso-mal fusion patterns between the 1q donor chromosomeand different recipient chromosomes. However, all studiesdone by routine chromosome analysis lacked the molecularcharacterization of breakpoints involving the chromosomefusion, underlying genomic imbalances, and tumor genemutations. A recent study using genomic array testing andtargeted gene panel next-generation sequencing detected the100–107 Mb duplication of 1q with breakpoints in 1q21.1-q21.3 region and recurrent mutations in the TET2 and SF3B1genes [10]. The TET2 gene encodes a methylcytosine dioxy-genase that catalyzes the conversion of methylcytosine to 5-hydroxymethylcytosine. Mutations in this gene have beenassociated with MDS (OMIM#612839) and responsivenessto hypomethylating agents such as azacitidine or decitabine[10]. The SF3B1 gene encodes subunit 1 of the splicingfactor 3b protein complex. Splicing factor 3b, together withsplicing factor 3a and a 12S RNA unit, forms the U2 smallnuclear ribonucleoproteins complex (U2 snRNP). Mutationsin the SF3B1 gene have been seen in MDS (OMIM#614286).These findings supported further genomic analysis to definethe breakpoints involving the jumping translocations anddisease-related gene mutations [12, 13].

Approximately one-third of MDS/AML cases with jump-ing translocations of 1q either emerged from a stemlineabnormal clone or evolved additional clonal abnormalities

Page 3: Jumping Translocations of 1q in Myelodysplastic Syndrome and … · 2019. 7. 30. · 9 MDS 8 1q12 2 10 MDS 8 1q31 2 11 MDS 8 1q12 2 12 MDS 8 1q12 4 13 MDS 8 1q12 2 14 MDS 8 1q21 5

Case Reports in Genetics 3

Case 1

Case 2

Case 3

der(14)t(1;14) dup(1q)der(21)t(1;21)

der(15)t(1;15) der(15)t(1;15),del(20q)

der(22)t(1;22) der(15)t(1;15),del(11q)der(15)t(1;15)

der(15)t(1;15) der(Y)t(Y;1),t(10;17)der(22)t(1;22)

(a)

Hypomethylation

Loss of Telomeric Function

Normal triradialBrokenTriradialDecondensedHypomethylatedChr 1 1q12

Normal Donor-recipientShortenedVariant telomereChr repeats translocationsfusiontelomere

Jumping

1q12

(b)

Figure 1: (a) Jumping translocations of 1q observed in the three patients.The jumping translocation in each cell lineage is shown inside a circle.Arrow points to an additional rearrangement evolved in a cell lineage. (b) Epigenetic alterations in the formation of jumping translocations of1q. Hypomethylation of 1q12 pericentric heterochromatin induced chromatin decondensation, triradial formation, and break-off of 1q donorsegment. Loss of telomeric function by variant telomeric repeats and shortened telomeres produced instable recipient chromosomes. Thesuccessive fusions between break-off donor segment and recipient chromosomes resulted in clonal abnormalities with cell linages sharinggains of donor chromosome segment.

Recipient chromosomes with fusion at pter or qter Recipient chromosomes with fusion at pericentric/interstitial regions

No. MDS/AML Refs Other Abnormalities (sl. stemline) Donor 1p 2q 3q 4p 4q 5p 5q 7p 8p 8q 9q 10q 11q 12p 12q 13p 13q 14p 14q 15p 16q 17q 18q 19p 19q 21p 21q 22p 22q Xq Yq 1q 5p12 6p226q12q14 7q11 8p21 12q13 15q11

16q11.2q12

20q12q13.1 Xp22.1

No, Cell Lineages

1 AML (M5b) 1 1q11 42 AML (M0) 2 51,XX,+6,+8,+9,+19,+20[sl] 1q21 33 MDS 3 47,XX,+8[sl] 1q11 34 MDS 3 46,XY,t(4;11)(q10;p15)[sl] 1q11 25 MDS > AML 4 46,XX,der(7)t(1;7)(q21;p22)[sl] 1q21 96 AML (M5b) 5 47,XY,+8[sl] 1q31q32 57 AML (M5a) 6 46,X,-Y,+8[sl] 1q12 38 AML (M1) 7 46,XX,t(1;6)(p32;q25-q27),

der(7)t(7;12)(q11.2~22;q13~q15)1q11 4

9 MDS 8 1q12 210 MDS 8 1q31 211 MDS 8 1q12 212 MDS 8 1q12 413 MDS 8 1q12 214 MDS 8 1q21 515 MDS 8 1q21 216 MDS 8 1q21 217 MDS 8 1q21 218 MDS 8 1q12 219 MDS 8 1q12 420 MDS 8 1q21 421 MDS 8 1q12 1022 MDS 8 1q11 223 MDS 8 1q11 324 MDS 8 1q12 225 AML 9 1q12 226 MDS 10 1q21 527 MDS 10 45,XX,der(13;15)(q10;q10) 1q12 428 MDS 10 1q21 229 MDS 10 46,Y,der(X)t(X;1)(q28;q21),del(6)(p22) 1q21 230 MDS 10 46,XY,add(18)(p11.2)[sl]

/46,sdl,del(13)(q12q22),del(20)(q11.2q13.1)1q21 3

31 MDS > AML Case 1 46,XY,der(1;15)(q12;p12),del(20)(q11.2q13.3) 1q12 4

32 MDS > AML Case 2 46,XY,der(1;15)(q12;p12),del(11)(q13q23) 1q12 233 MDS > AML Case 3 46,X,der(Y)t(Y;1)(q12;q11),t(10;17)(p10;p10) 1q12 3

Count of Recipient Chromosomes::

1 1 1 2 1 1 2 1 1 1 1 1 1 1 1 4 2 8 1 13 1 4 5 1 2 9 3 12 2 1 4 2 1 1 2 3 2 1 1 4 3 1 110Relative Frequencies of Recipient Chromosomes: 4% 7% 12% 4% 5% 8% 3% 11% 4% 3% 4% 3%

*filled blue box for telomeric or subtelomeric fusion at recipient chromosomes, and filled orange box for pericentric or interstitial fusion at recipient chromosomes.

Figure 2: Patterns of donor and recipient chromosomes in jumping translocations of 1q of MDS and AML patients.

and two-thirds were detected as de novo chromosomalabnormalities. Shortened telomeres and many variant telom-eric repeats were detected in the fusion points from jumpingtranslocations of 1q in a case ofMDS progressed to AML; thisresult suggested that the extended proliferation of cancer cells

might be related to the loss of telomeric function [4]. Copynumber gain of 1q involving pericentromeric breakpointshad been seen frequently in hepatocellular carcinoma. It waspostulated that hypomethylation of satellite 2 DNA at 1q12alters the interaction between the CpG-rich satellite DNA

Page 4: Jumping Translocations of 1q in Myelodysplastic Syndrome and … · 2019. 7. 30. · 9 MDS 8 1q12 2 10 MDS 8 1q31 2 11 MDS 8 1q12 2 12 MDS 8 1q12 4 13 MDS 8 1q12 2 14 MDS 8 1q21 5

4 Case Reports in Genetics

and chromatin proteins which results in heterochromatindecondensation, breakage, and aberrant 1q formation [14].Investigation of der(16)t(1;16)/der(1;16) in breast carcinomassuggested that hypomethylation of pericentromeric satellite2 DNA in 1q12 might be associated with the accumulationof many numerical chromosome alterations and aggressivehistological features [15]. The study of five patients with1q12 rearrangements causingmultiplemyeloma observed thathypomethylation of the 1q12 pericentromeric heterochro-matin induced transient decondensations and triradials ofthe 1q12 pericentromeric region. Triradials of 1q12 originatedclonal unbalanced 1q rearrangements or jumping transloca-tions by successive fusing to the telomeric or pericentromericregions of recipient chromosomes [16]. Therefore, the pres-ence of jumping translocations indicated chromosomal insta-bility by altered epigenetic modifications. A proposedmecha-nism for the formation of jumping translocations is schemedin Figure 1(b).

Theprogression ofMDS toAMLandpoor prognosiswerenoted in the present three cases with jumping translocationsof 1q. This result was consistent with previous findings inwhich jumping translocations of 1q were associated witha high risk of progression to AML and poor prognosisand thus warranted aggressive therapy [8, 10]. Currently,the azanucleosides azacitidine and decitabine are used forthe treatment of MDS and AML [17]. These azanucleosidesare referred to as hypomethylating agents which interferewith the DNA methylation machinery. The incorporationof azacytidine into RNA or decitabine into DNA triggerstwo main antitumor cellular activities through the inductionof cytotoxicity leading to DNA damage response and DNAhypomethylation through inhibition of DNA methyltrans-ferase which enables reactivation of tumor suppressor genesfor normal growth and differentiation. For MDS and AMLwith jumping translocations of 1q induced by hypomethy-lation at 1q pericentric heterochromatin, the treatment byazanucleosides may rely on the cytotoxicity effect. Furtherstudies to understand the underlying genomic defects causingjumping translocations andmechanism of action for targetedtreatment are needed.

In summary, jumping translocations of 1q have beenshown as an indication of chromosomal instability likelyinduced by epigenetic alterations of hypomethylation andshortened telomeres. The jumping translocations of 1qshowed unique rearrangement patterns with multiple celllineages which all have a gain of donor chromosome segmentand lack other complex structural or numerical rearrange-ments. It is not clear whether these epigenetic alterationsoccur independently as a two-step event or concurrently bydriving genetic defects. Further molecular analyses to under-stand the genetic defects causing jumping translocations of 1qand related disease-causing mechanism can facilitate targetedtherapy for this type of MDS and AML.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

[1] A. Asahara, E. Morioka, S. Hisano et al., “A case of acutemonocytic leukemia with chromosome abnormality, jumpingtranslocation,” Japanese Journal Clinical Hematology, vol. 32, p.1244, 1991.

[2] T. Shikano, H. Arioka, R. Kobayashi, H. Naito, and Y. Ishikawa,“Jumping translocations of 1q in Burkitt lymphoma and acutenonlymphocytic leukemia,” Cancer Genetics and Cytogenetics,vol. 71, no. 1, pp. 22–26, 1993.

[3] V. Najfeld, B. Hauschildt, A. Scalise et al., “Jumping transloca-tions in leukemia,” Leukemia, vol. 9, no. 4, pp. 634–639, 1995.

[4] S. Hatakeyama, K. Fujita, H. Mori, M. Omine, and F. Ishikawa,“Shortened telomeres involved in a casewith a jumping translo-cation at 1q21,” Blood, vol. 91, no. 5, pp. 1514–1519, 1998.

[5] K. Reddy, L. Parsons, and L. Colman, “Jumping TranslocationsInvolving Chromosome 1q in a Patient with Crohn Disease andAcuteMonocytic Leukemia,” Cancer Genetics and Cytogenetics,vol. 109, no. 2, pp. 144–149, 1999.

[6] M. Busson Le Coniat, F. Brizard, N. V. Smadja, O. Maarek, H.Der Sarkissian, and R. Berger, “Interstitial telomere repeats intranslocations of hematopoietic disorders,” Leukemia, vol. 14,no. 9, pp. 1630–1633, 2000.

[7] K.N.Manola, V.N.Georgakakos, C. Stavropoulou et al., “Jump-ing translocations in hematological malignancies: a cytogeneticstudy of five cases,” Cancer Genetics and Cytogenetics, vol. 187,no. 2, pp. 85–94, 2008.

[8] V. Najfeld, J. Tripodi, A. Scalise et al., “Jumping translocationsof the long arms of chromosome 1 in myeloid malignancies isassociated with a high risk of transformation to acute myeloidleukaemia,” British Journal of Haematology, vol. 151, no. 3, pp.288–291, 2010.

[9] M. Parihar, A. Gupta, A. K. Yadav, D. K. Mishra, A. Bhat-tacharyya, and M. Chandy, “Jumping translocation in a caseof de novo infant acute myeloid leukemia,” Pediatric Blood &Cancer, vol. 61, no. 2, pp. 387–389, 2014.

[10] C. C. S. Yeung, H. J. Deeg, C. Pritchard, D. Wu, and M. Fang,“Jumping translocations inmyelodysplastic syndromes,”CancerGenetics, vol. 209, no. 9, pp. 395–402, 2016.

[11] E. Bourneuf, F. Herault, C. Chicault et al., “Microarray analysisof differential gene expression in the liver of lean and fatchickens,”Gene, vol. 372, no. 1-2, pp. 162–170, 2006.

[12] R. Bajaj, F. Xu, B. Xiang et al., “Evidence-based genomicdiagnosis characterized chromosomal and cryptic imbalancesin 30 elderly patients with myelodysplastic syndrome and acutemyeloid leukemia,” Molecular Cytogenetics, vol. 4, no. 1, p. 3,2011.

[13] S. D. Hudnall, H. Meng, L. Lozovatsky, P. Li, M. Strout, and S.H. Kleinstein, “Recurrent genetic defects in classical Hodgkinlymphoma cell lines,” Leukemia & Lymphoma, vol. 57, no. 12,pp. 2890–2900, 2016.

[14] N. Wong, W.-C. Lam, P. B.-S. Lai, E. Pang, W.-Y. Lau, and P. J.Johnson, “Hypomethylation of chromosome 1 heterochromatinDNA correlates with q-arm copy gain in human hepatocellularcarcinoma,” The American Journal of Pathology, vol. 159, no. 2,pp. 465–471, 2001.

[15] H. Tsuda, T. Takarabe, Y. Kanai, T. Fukutomi, and S. Hirohashi,“Correlation of DNAhypomethylation at pericentromeric hete-rochromatin regions of chromosomes 16 and 1 with histologicalfeatures and chromosomal abnormalities of human breastcarcinomas,”The American Journal of Pathology, vol. 161, no. 3,pp. 859–866, 2002.

Page 5: Jumping Translocations of 1q in Myelodysplastic Syndrome and … · 2019. 7. 30. · 9 MDS 8 1q12 2 10 MDS 8 1q31 2 11 MDS 8 1q12 2 12 MDS 8 1q12 4 13 MDS 8 1q12 2 14 MDS 8 1q21 5

Case Reports in Genetics 5

[16] J. R. Sawyer, E. Tian,C. J.Heuck et al., “Evidence of an epigeneticorigin for high-risk 1q21 copy number aberrations in multiplemyeloma,” Blood, vol. 125, no. 24, pp. 3756–3759, 2015.

[17] J. Diesch, A. Zwick, A.-K. Garz, A. Palau, M. Buschbeck, and K.S. Gotze, “A clinical-molecular update on azanucleoside-basedtherapy for the treatment of hematologic cancers,” ClinicalEpigenetics, vol. 8, no. 1, 2016.

Page 6: Jumping Translocations of 1q in Myelodysplastic Syndrome and … · 2019. 7. 30. · 9 MDS 8 1q12 2 10 MDS 8 1q31 2 11 MDS 8 1q12 2 12 MDS 8 1q12 4 13 MDS 8 1q12 2 14 MDS 8 1q21 5

Stem Cells International

Hindawiwww.hindawi.com Volume 2018

Hindawiwww.hindawi.com Volume 2018

MEDIATORSINFLAMMATION

of

EndocrinologyInternational Journal of

Hindawiwww.hindawi.com Volume 2018

Hindawiwww.hindawi.com Volume 2018

Disease Markers

Hindawiwww.hindawi.com Volume 2018

BioMed Research International

OncologyJournal of

Hindawiwww.hindawi.com Volume 2013

Hindawiwww.hindawi.com Volume 2018

Oxidative Medicine and Cellular Longevity

Hindawiwww.hindawi.com Volume 2018

PPAR Research

Hindawi Publishing Corporation http://www.hindawi.com Volume 2013Hindawiwww.hindawi.com

The Scientific World Journal

Volume 2018

Immunology ResearchHindawiwww.hindawi.com Volume 2018

Journal of

ObesityJournal of

Hindawiwww.hindawi.com Volume 2018

Hindawiwww.hindawi.com Volume 2018

Computational and Mathematical Methods in Medicine

Hindawiwww.hindawi.com Volume 2018

Behavioural Neurology

OphthalmologyJournal of

Hindawiwww.hindawi.com Volume 2018

Diabetes ResearchJournal of

Hindawiwww.hindawi.com Volume 2018

Hindawiwww.hindawi.com Volume 2018

Research and TreatmentAIDS

Hindawiwww.hindawi.com Volume 2018

Gastroenterology Research and Practice

Hindawiwww.hindawi.com Volume 2018

Parkinson’s Disease

Evidence-Based Complementary andAlternative Medicine

Volume 2018Hindawiwww.hindawi.com

Submit your manuscripts atwww.hindawi.com