Skip to main content

ORIGINAL RESEARCH article

Front. Oncol., 30 May 2018
Sec. Molecular and Cellular Oncology
This article is part of the Research Topic Cancer Models View all 14 articles

Genome-Wide Analysis of Interchromosomal Interaction Probabilities Reveals Chained Translocations and Overrepresentation of Translocation Breakpoints in Genes in a Cutaneous T-Cell Lymphoma Cell Line

\r\nAnne Steininger&#x;Anne Steininger1†Grit Ebert&#x;Grit Ebert1†Benjamin V. BeckerBenjamin V. Becker2Chalid AssafChalid Assaf3Markus MbsMarkus Möbs4Christian A. SchmidtChristian A. Schmidt5Piotr GrabarczykPiotr Grabarczyk5Lars R. JensenLars R. Jensen6Grzegorz K. PrzybylskiGrzegorz K. Przybylski7Matthias PortMatthias Port2Andreas W. KussAndreas W. Kuss6Reinhard Ullmann*\r\nReinhard Ullmann2*
  • 1Max Planck Institute for Molecular Genetics, Berlin, Germany
  • 2Bundeswehr Institute of Radiobiology Affiliated to the University of Ulm, Munich, Germany
  • 3Department of Dermatology and Venerology, Helios Klinikum Krefeld, Krefeld, Germany
  • 4Berlin Institute of Health, Institute of Pathology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
  • 5Clinic for Internal Medicine C, University Medicine Greifswald, Greifswald, Germany
  • 6Human Molecular Genetics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
  • 7Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland

In classical models of tumorigenesis, the accumulation of tumor promoting chromosomal aberrations is described as a gradual process. Next-generation sequencing-based methods have recently revealed complex patterns of chromosomal aberrations, which are beyond explanation by these classical models of karyotypic evolution of tumor genomes. Thus, the term chromothripsis has been introduced to describe a phenomenon, where temporarily and spatially confined genomic instability results in dramatic chromosomal rearrangements limited to segments of one or a few chromosomes. Simultaneously arising and misrepaired DNA double-strand breaks are also the cause of another phenomenon called chromoplexy, which is characterized by the presence of chained translocations and interlinking deletion bridges involving several chromosomes. In this study, we demonstrate the genome-wide identification of chromosomal translocations based on the analysis of translocation-associated changes in spatial proximities of chromosome territories on the example of the cutaneous T-cell lymphoma cell line Se-Ax. We have used alterations of intra- and interchromosomal interaction probabilities as detected by genome-wide chromosome conformation capture (Hi-C) to infer the presence of translocations and to fine-map their breakpoints. The outcome of this analysis was subsequently compared to datasets on DNA copy number alterations and gene expression. The presence of chained translocations within the Se-Ax genome, partly connected by intervening deletion bridges, indicates a role of chromoplexy in the etiology of this cutaneous T-cell lymphoma. Notably, translocation breakpoints were significantly overrepresented in genes, which highlight gene-associated biological processes like transcription or other gene characteristics as a possible cause of the observed complex rearrangements. Given the relevance of chromosomal aberrations for basic and translational research, genome-wide high-resolution analysis of structural chromosomal aberrations will gain increasing importance.

Introduction

The analysis of structural chromosomal aberrations is of relevance for both basic and translational research. Several chromosomal markers are already routinely used in clinical tests for genotype-based sub-classification of tumors or to assist in therapeutic decisions. In addition, the identification of recurrent aberrations can highlight driver genes of tumorigenesis, which represent promising starting points for the development of targeted therapies. Apart from clinical applications, the characterization of chromosomal aberrations can shed light on the underlying mutational mechanisms and in this way contribute to a better understanding of the cause and course of intra-individual evolution of tumors.

According to classical models of tumorigenesis, complex abnormal karyotypes emerge through the stepwise acquisition of chromosomal rearrangements followed by expansion of those mutated clones with highest proliferative capacity (1). Yet, the conception of intra-individual karyotypic evolution has been biased by the limited perspective as provided by the low resolution of genome-wide datasets on structural chromosomal rearrangements for a long time. The lack of appropriate techniques capable of capturing karyotypic complexity both genome-wide and with high resolution has hampered the identification of mechanisms alternative to the well-documented gradual process. The introduction of array-based comparative genomic hybridization [(arrayCGH) (2, 3)] has mitigated this technical shortcoming for unbalanced structural chromosomal aberrations, but the situation has remained unsatisfactory for balanced chromosomal rearrangements. Until recently, their characterization required time-consuming cloning of breakpoints or, in case of translocations, depended on sophisticated sorting of derivative chromosomes followed by hybridization of sorted chromosomes on DNA microarrays (46). This situation has changed with the advent of next-generation sequencing (NGS), which has set the stage for the development of new protocols for the analysis of structural chromosome aberrations (7). Initially, these analyses have mainly focused on alterations of sequencing depth across the genome or along sorted chromosomes to define DNA copy number changes and translocation breakpoints (8), respectively. Later protocols have taken advantage of paired-end reads and used their mapping position and orientation with respect to the human reference genome to infer the presence and location of structural chromosome aberrations (9). Despite the development of paired-end NGS protocols, the identification of structural chromosomal aberrations such as balanced translocations has remained challenging. This is mainly due to the fact that strategies based on standard paired-end sequencing protocols have to rely on those few sequenced chimeric fragments that span the chromosomal breakpoints. Hence, reliable detection of such rearrangements using standard NGS protocols requires considerable sequencing depth (10). Furthermore, even in case of sufficient sequencing depth, translocation breakpoints in the very vicinity of regions with low mappability, such as repetitive elements, segmental duplications or DNA segments with extreme bias of base composition might be missed or erroneously aligned (11). An alternative strategy capable to overcome these problems is based on the fact that chromosomal rearrangements such as translocations disrupt nuclear architecture and modify spatial proximities of chromosome territories. These modifications of nuclear organization can be monitored by chromosome conformation capture assays such as Hi-C (1214). This technique combines proximity ligation and NGS to infer nuclear neighborhood of chromosomal regions (see Figure 1 for explanation). The closer two chromosomal segments are within the nucleus, the more frequent Hi-C will detect interactions between them.

FIGURE 1
www.frontiersin.org

Figure 1. Principle of Hi-C. Hi-C is a variant of the chromosome conformation capture assay dedicated to the identification of genome-wide intrachromosomal and interchromosomal interaction probabilities. The method starts with crosslinking of chromatin within the nuclei. Restriction enzyme digestion (in this case HindIII) generates sticky ends which are filled-in and labeled with biotin. During the following ligation step, free DNA ends are re-ligated, which can either restore the original DNA sequence or can lead to chimeric products. The likelihood of such chimeric products depends on their spatial proximity. Afterward, crosslinking is reversed and DNA is sheared. Size selection and pull-down of biotin-labeled fragments are performed to increase specificity. Next, sequencing adapters are ligated to each fragment before sequencing. Finally, paired-end sequences are mapped to the human reference genome to deduce spatial proximities from the frequency of chimeric sequences.

In general, interaction frequencies between two regions on the same chromosome decrease with linear distance and interactions between two chromosomes are considerably rare when compared to intra-chromosomal ones. In case of a translocation, regions of two or more chromosomes come into close contact. This results in an abrupt increase of interaction frequencies between the segments adjacent to the translocation breakpoints and makes Hi-C an ideal approach for the detection of balanced translocations (10, 1517).

The application of array- and sequencing-based approaches as described above unveiled an unprecedented complexity of structural chromosomal aberrations in tumor genomes. In several cases, the observed mutational patterns were hardly compatible with a stepwise accumulation of chromosomal aberrations as described by the classical model of tumorigenesis (18). For example, regionally confined clusters of numerous chromosomal aberrations with limited DNA copy number states in the absence of general genomic instability have suggested a single catastrophic event as the underlying cause of this complex pattern of aberrations instead of a series of consecutive events. Meanwhile, it has been shown that this phenomenon, termed chromothripsis, can be encountered in a broad range of tumor types, where it can affect 2–3% of patients (19). In 2015, Zhang and colleagues succeeded to demonstrate that micronuclei formation and DNA damage confined to these structures can produce similar patterns of chromosomal changes as typical for chromothripsis (20). Multiple simultaneously arising DNA double-strand breaks also account for the emergence of chromoplexy, which is characterized by chained translocations and interlinking deletion bridges involving numerous chromosomes (21). The complex patterns of chromosomal aberrations typical for chromoplexy are unlikely result of a stepwise process, as this would require the repeated use of the same chromosomal breakpoints (18, 21). It is still unclear what triggers these simultaneously arising DNA strand breaks in the context of chromoplexy (18).

In this study, we have employed chromosomal interaction probabilities to fine-map translocations in a cell line derived from a patient with Sézary syndrome. The etiology of this highly malignant cutaneous T-cell lymphoma is poorly understood (22) and thorough investigation of structural chromosomal aberrations promises more insights into its development and progression (23). We demonstrate the presence of chained translocations partly connected by interlinking deletion bridges, which suggests the manifestation of chromoplexy and argues against a gradual appearance of these chromosomal aberrations. The overrepresentation of chromosomal translocation breakpoints within genes highlights the possible impact of spatial proximities of genes and biological processes associated with genes on the emergence of chromosomal translocations in this cutaneous T-cell lymphoma.

Materials and Methods

Hi-C is a high-throughput variant of the chromosome conformation capture assay and facilitates the genome-wide investigation of interaction probabilities of genomic segments within the nucleus. The technology is based on crosslinking of chromatin, fragmentation of DNA followed by re-ligation. Depending on their spatial distribution, not only the original DNA fragments but also fragments in spatial proximity will re-ligate. These chimeric fragments can be detected by paired-end sequencing and their frequency can be used to calculate the interaction probability of these fragments within the nucleus. The principle of Hi-C is schematically depicted in Figure 1. For this study, we have used a Hi-C protocol published in detail by Lieberman-Aiden and colleagues (13).

Fixation, Cell Lysis, and Restriction Enzyme Digestion

In brief, 20–25 million Se-Ax cells (24) were cross-linked with formaldehyde (Thermo Fisher Scientific, Waltham, MA, USA). Crosslinking was stopped by addition of 125 mM glycine (Merck Millipore, Darmstadt, Germany). After washing cells in ice-cold DPBS buffer (Lonza, Basel, Switzerland), cell pellets were flash frozen and stored at −80°C. For lysis, cells were resuspended in Hi-C lysis buffer and lysed using a Dounce homogenizer (Fisher Scientific GmbH, Schwerte, Germany). After centrifugation, pellets were washed twice in NEB buffer 2 (New England Biolabs, Ipswhich, MA, USA), finally resuspended in 370 µl NEB buffer 2 and 50 µl were transferred to seven tubes each. In order to remove proteins not cross-linked to DNA, 38 µl 1%SDS (Sigma-Aldrich, St. Louis, MO, USA) was added to each tube and incubated for 10 min at 65°C. Afterward, SDS was inactivated by the addition of 44 µl 10% Triton X-100 (Sigma-Aldrich, St. Louis, MO, USA). In each but one tube 400 U HindIII (New England Biolabs, Ipswhich, MA, USA) were added and DNA was digested overnight at 37°C with rotation. The next day, the tube with undigested DNA and one HindIII treated sample were removed to verify HindIII digestion efficiency.

Endlabeling, Re-Ligation, Reversal of Crosslinking, and DNA Purification

For the remaining tubes a fill-in reaction was performed to blunt the sticky ends as generated by HindIII digestion. For later enrichment of re-ligated fragments, Biotin-dCTP (Invitrogen, Carlsbad, CA, USA) was incorporated during this fill-in reaction. Fragments were re-ligated with 15 U T4 DNA ligase per tube for 4 h at 16°C. Reversal of crosslinking was made by addition of 25 µl Proteinase K (20 mg/ml, Thermo Fisher Scientific, Waltham, MA, USA) and incubation overnight at 65°C. After RNA digestion with 50 µl RNase A (10 mg/ml, Thermo Fisher Scientific, Waltham, MA, USA) for 45 min at 37°C, DNA was purified by means of standard phenol chroroform isoamylalcohol treatment (Sigma-Aldrich, St. Louis, MO, USA) and ethanol (Merck Millipore, Darmstadt, Germany) precipitation. DNA of the separate tubes was conflated and concentration measured with a Qubit fluorometric assay (Thermo Fisher Scientific, Waltham, MA, USA).

Removal of Biotin From Unligated DNA Ends, Enrichment of Re-Ligated Fragments and Preparation of Sequencing Libraries

In order to remove biotin-labeling from unligated fragments, samples were treated for 2 h at 12°C with 5 U T4 DNA polymerase (New England Biolabs, Ipswhich, MA, USA), whose exonuclease activity removed the biotin at the ends of the unligated fragments while keeping the centrally positioned biotin of ligated fragments untouched. Library generation was done according to the manufacturer’s protocols, with minor adaptions concerning the pull-down of biotin-labeled fragments to eliminate unligated fragments. In brief, 5 µg DNA was sheared with the Covaris S2 system (Covaris, Woburn, MA, USA), DNA end-repaired and size selected by means of Agencourt AMPure XP Reagent beads (Beckman Coulter Genomics, Danvers, MA, USA). Afterward, size and quantity was verified employing a 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA). Unligated fragments were depleted by pull-down of biotin-labeled fragments with 50 µl streptavidin beads (10 mg/ml, Life Technologies, Carlsbad, CA, USA). The resulting DNA-coated beads were resuspended in 34 µl TE buffer (Life Technologies, Carlsbad, CA, USA). After A-tailing, i.e., addition of a dATP to the repaired DNA ends, and ligation of sequencing adapters, 5 µl of DNA-coated beads were used for 10 cycles of PCR amplification with primers complementary to the ligated sequencing adapters. These amplicons were sequenced using the SOLiD 5500X1 Sequencing Instrument (Life Technologies, Carlsbad, CA, USA) using the paired-end protocol with 75 and 35 nucleotides for the forward and reverse strand, respectively.

Processing and Quality Control of Hi-C Data

Forward and reverse sequence reads were separately aligned to the human reference genome (hg19) by means of LifeScope Genomic Analysis Software 2.5.1. The resulting file was imported into the software tool HOMER v.4.7 (25), where the dataset was filtered for possible PCR artifacts, reads with low mapping quality and reads derived from sites lacking HindIII motifs. For visualization of chromosomal interaction probabilities as heatmaps, reads that passed the above-mentioned filters were summarized to genomic bins of 100 and 250 kb in size and read counts were normalized. The normalization strategy as implemented in HOMER proceeds on the assumption that each region within the genome should have the same visibility and for that reason equalizes possible artifactual effects caused by differences in GC content, accessibility of DNA and unequal distribution of HindIII sites. Significantly interacting genomic bins were determined employing HOMER’s analyzeHiC module (FDR = 0.001; bin interaction distance >25 Mb). Visualization of interaction frequencies in heatmaps was done in JAVA Treeview (26). Translocations were preselected by visual inspection of the interaction heatmaps. Translocation breakpoints were fine-mapped by evaluating read distribution within a 2 Mb window surrounding the breakpoints in order to identify the HindIII fragment next to the breakpoint.

Comparison of Translocation Breakpoint Regions With Data on Higher Order Chromatin Conformation, DNA Copy Number Alterations, and Search for Expressed Fusion Genes

Given the hypothesis that spatial proximity might promote the emergence of translocations, we have processed public Hi-C data on the B-lymphocyte cell line GM12878 (27) in the same ways as the data for Se-Ax and evaluated the presence of significant interchromosomal interactions as defined by HOMER (FDR = 0.001) connecting the translocation partner chromosomes by means of Circos (28). Additionally, we have visually inspected various public data on chromatin interaction deposited at the 4DGenome database (29) (https://4dgenome.research.chop.edu/) to identify possible interactions between our intervals of interest in other cell lines.

Data on DNA copy number alterations in Se-Ax that have been generated by means of arrayCGH in a previous study (30) were visualized for each translocation within a 2 Mb interval surrounding the breakpoint by means of R and the R packages reshape2 (31) and ggplot2 (32). The expression of fusion genes was tested using previously published RNA-Seq data (33, 34). Translocation breakpoints were verified by screening paired-end sequencing data for Se-Ax (33, 34) and the analysis of these data by Breakdancer (35).

Analysis of Chromosomal Breakpoint Overrepresentation Within Genes

Overrepresentation of translocation breakpoints within genes was tested at the resolution of single HindIII fragments by calculating the likelihood that the same number of randomly distributed HindIII fragments map to genes as it has been observed for HindIII fragments located next to the translocation breakpoints. In a first step, we have cataloged all HindIII restriction sites within the human genome by means of Galaxy Emboss command fuzznuc (36, 37). Gaps in the human genome assembly (38) were subtracted with BEDtools (39). From the resulting dataset, the Unix command shuf was employed to generate 100,000 permutations of 32 HindIII fragments and the BEDtools command “intersectBed” was used to compute the frequency of overlap with RefSeq genes (40).

To calculate the p-value for Monte Carlo resampling according to Ref. (41), the number of permutation datasets that feature an equal or greater count of HindIII fragment regions with gene overlap as observed (> = 24) were used as the expected overlap.

Results

Hi-C analysis was based on 91.9 million read pairs that passed processing and quality filtering in HOMER. A genome-wide survey of structural aberrations is presented in Figure 2. This heatmap depicts the ratio of observed interaction frequencies and the expected frequencies based on a background model. Translocations are indicated by higher than expected frequencies of interchromosomal interactions (red color). Correspondingly, intrachromosomal interaction frequencies of the chromosomes involved in the translocation are decreased (blue color). The color gradient indicates the orientation of the breakpoint; i.e., interaction intensities decrease with distance from chromosomal breakpoints. In total, we identified 22 translocations, from which we were able to fine-map 32 breakpoints to a single HindIII fragment (Table 1; note that only 32 of the 34 breakpoints as listed in Table 1 were considered for the following analysis as in two cases breakpoints mapped to the same HindIII fragment). A comparison of Hi-C data with whole-genome sequencing data generated by a different laboratory using a different batch of Se-Ax cells (33, 34) revealed an overlap of 25 breakpoints. These have been highlighted in Table 1. A comparison of translocation breakpoints with array CGH data generated in a previous study by our laboratory with a resolution of ~100 kb (30) revealed that 11 of those breakpoints not identified by whole-genome sequencing were flanked by either deletions (n = 7) or duplications (n = 4). Other translocation breakpoints solely identified by Hi-C analysis were in close vicinity to other translocations, suggesting the presence of a complex rearrangement (t1/t10; t7/t8; t8/t15; and t13/t14). Yet, it has to be emphasized that non-overlapping breakpoints may also be owed to private mutations emerging during cultivation of Se-Ax cells in different laboratories over longer time or other technical reasons, in particular differences in resolution.

FIGURE 2
www.frontiersin.org

Figure 2. Genome-wide interaction frequencies in Se-Ax. Higher and lower than expected normalized interaction frequencies are shown with 2.5 Mb resolution in red and blue, respectively. The chromosome numbers are given at the top and to the right; together with information on DNA copy number losses (red) and gains (green) as detected by array comparative genomic hybridization. Translocations are characterized by interchromosomal interactions higher than expected, while their corresponding intrachromosomal interactions are decreased. A more detailed view of selected chromosomes is provided in Figure 3.

TABLE 1
www.frontiersin.org

Table 1. Translocation breakpoints (hg19).

As an example for the complexity of chromosomal aberrations, a zoom-in depicting interchromosomal interactions for chromosomes 2, 6, and 11 is given in Figure 3. Additionally, chromosomal deletions and duplications identified by arrayCGH analysis of Se-Ax are indicated in both heatmaps.

FIGURE 3
www.frontiersin.org

Figure 3. Heatmap of normalized interchromosomal interaction frequencies between chromosomes 2 and 6 and chromosomes 2 and 11. Two heatmaps are shown, which demonstrate the presence of a translocation t(2;6) (left) and t(2;11) (right), respectively. Both derivative chromosomes lead to higher than expected interchromosomal interaction frequencies, which are indicated by the red color gradient. Alterations of DNA copy number state as detected by array comparative genomic hybridization is indicated by coloring of the chromosome ideograms (red = deletion, green = gain). While the breakpoint of reciprocal translocation t(2;11) is easily identifiable [1], the identification of t(2;6) [2] is complicated by additional deletions of chromosome 2 [3] and chromosome 6 [4] and an inversion of chromosome 2 [5]. Orientation of chromosomal rearrangements can be inferred from the color gradient [interaction intensities (i.e., red color) decrease with distance from chromosomal breakpoints].

Deletions adjacent to transclocation breakpoints have been encountered 12 times (out of 32 breakpoints; Figure 4). The Circos plot depicted in Figure 5 demonstrates chained translocations with shared breakpoints between several chromosomes on the example of chromosome 5, 8, and 10.

FIGURE 4
www.frontiersin.org

Figure 4. Deletions adjacent to the translocation breakpoints identified in Se-Ax. Smoothed log2 ratios of DNA copy number within a 2 Mb interval surrounding the translocation breakpoints are shown for Se-Ax (red line). DNA copy numbers of additional cell lines for the very same intervals are displayed for comparison (see insert box for color legend).

FIGURE 5
www.frontiersin.org

Figure 5. Circos plot visualizing chained translocations between chromosomes 5, 8 and 10. In this Circos plot chromosomes are radially aligned. Arcs within this circle indicate significant interchromosomal and long distance intrachromosomal interactions. Following the numbering given in the small insert to the left: (1) significant interchromosomal interactions (blue lines; FDR <0.001), significant long distance intrachromosomal interactions (gray lines; >25 Mb, FDR <0.001) and translocations as given in Table 1 (black lines); (2) radially aligned chromosome ideograms; (3) count of significant interactions per 50 kb bin (all interaction distances; max = 10); (4) DNA copy number status in red (deletion) and green (gain) as detected by array comparative genomic hybridization.

In order to evaluate the impact of spatial proximity of chromosomes on the emergence of translocations, we screened public Hi-C datasets for interactions between those chromosomal intervals affected by translocations in Se-Ax. We failed to get any clues on higher interaction probabilities between regions encompassing the translocation regions, neither in the data of the lymphoblastoid cell line GM12878, which we processed the same way as the Se-Ax data, nor in the datasets from the 4DGenome database. Permutation analysis revealed a significant overrepresentation of translocation breakpoint-associated HindIII fragments within genes (p = 0.00208, 100,000 permutations). For one of the possible fusion genes (AIG1/GOSR1), transcripts were identified in the corresponding published RNA-Seq data (33).

Discussion

The genome-wide identification and fine-mapping of chromosomal aberrations in tumor cells is instrumental in getting insights into the molecular mechanisms underlying their formation. On the example of a cutaneous T-cell lymphoma cell line we demonstrated the usefulness of Hi-C for the identification of balanced chromosomal translocations. We could show that the abrupt and prominent change of chromosomal interaction probabilities caused by derivative chromosomes facilitates the identification of translocation partners, their orientation to each other as well as their chromosomal breakpoints. Thereby, the observed changes of interaction probabilities were not confined to the area surrounding the breakpoints, but have extended several megabases beyond (Figures 2 and 3). This makes the detection of translocations by Hi-C sensitive, robust, and less prone to artifacts, particularly if the chromosomal breakpoints are next to repetitive sequences, segmental duplications or DNA copy number aberrations. Consequently, the analysis of translocations by Hi-C overcomes some of the limitations of alternative deep sequencing approaches described above. Resolution and sensitivity of this approach might be further increased by recent modifications of the Hi-C protocol (27), by using more sequencing reads and more frequently cutting enzymes (e.g., 4 bp instead of 6 bp) (42) or DNase as an alternative (43). A current limitation of the presented approach is that the presence of translocations has been identified by visual inspection of interaction matrices and that their chromosomal breakpoints were pinned down to the level of single restriction enzyme recognition sites by scrutinizing read distribution within the preselected chromosomal intervals later on. There is need for automation and objectivation of this process and very recently, first software tools dedicated to this task have already been presented (44). Although the focus of this study is on balanced translocations, the observation of higher than expected interaction probabilities between chromosomal segments can also be employed for the detection of DNA copy number alterations and inversions, but particularly for small inversions higher sequencing depth is needed for their robust detection (10). Hi-C analysis with sufficient sequencing depth and read length would also facilitate the determination of haplotype phase (45, 46), which would allow the correct assignment of chromosomal breakpoints to either the maternally or paternally derived chromosomes.

In line with previous reports, the Hi-C data presented in this study have revealed a highly complex karyotype in the investigated cell line. Strikingly, several of the translocation breakpoints seem to be chained and associated with chromosomal deletions. Such patterns of rearrangements have already been observed in other tumors (47, 48), including one case of cutaneous T-cell lymphoma (49), and the term chromoplexy has been coined to describe this phenomenon (21). Simultaneously arising DNA double-strand breaks are a prerequisite for chromoplexy, but the triggers of the temporarily and spatially confined genomic instability have not been identified yet (18). Notably, the majority of breakpoints map within genes (p = 0.00208), which suggests that transcription or other biological processes associated with genic sequences could be involved in this mutational event (50). Irrespective of the cause of genomic instability the fusion of DNA double-strand breaks requires their spatial proximity, either before DNA damage (contact first) or thereafter, when broken ends might migrate to some sort of repair center (breakage first) (51). Although our comparison of translocation breakpoints with public chromatin interaction data did not produce any conclusive results, there is evidence in the literature that nuclear neighborhood of chromosomes impacts the frequency of translocations (52, 53). Against this background, transcription factories could be the possible scene of the observed punctuated accumulation of chromosomal translocations, as genes from different chromosomes cluster together in these nuclear structures (54, 55).

In summary, we have demonstrated the power of genome-wide chromosome conformation capture analysis to detect chromosomal translocations. We present evidence that several translocations identified in the investigated cutaneous T-cell lymphoma cell line likely emerged simultaneously leading to karyotypic features typical of chromoplexy. Overrepresentation of breakpoints within genic sequences highlights the role of transcription or gene-associated biological processes in the emergence of the observed pattern of structural chromosomal rearrangements.

Author Contributions

AS, GE, LRJ and AWK generated Hi-C data, which were analyzed by AS, GE and RU. GKP compared Hi-C results to whole genome sequencing data. CA, MM, GE, MP, CAS, BVB, PG LRJ, GKP and AWK contributed to data interpretation and critically reviewed the manuscript. AS and RU designed the study and wrote the manuscript.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

We thank Robert Weissmann for excellent bioinformatic support of Hi-C data.

Funding

This project was funded by the German Ministry of Defence. CA was funded by the Wilhelm-Sander Stiftung (2011-066.1) and GKP by the National Science Centre (decision No 2013/08/M/NZ2/00962).

References

1. Yates LR, Campbell PJ. Evolution of the cancer genome. Nat Rev Genet (2012) 13(11):795–806. doi:10.1038/nrg3317

CrossRef Full Text | Google Scholar

2. Solinas-Toldo S, Lampel S, Stilgenbauer S, Nickolenko J, Benner A, Dohner H, et al. Matrix-based comparative genomic hybridization: biochips to screen for genomic imbalances. Genes Chromosomes Cancer (1997) 20(4):399–407. doi:10.1002/(SICI)1098-2264(199712)20:4<399::AID-GCC12>3.0.CO;2-I

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Pinkel D, Segraves R, Sudar D, Clark S, Poole I, Kowbel D, et al. High resolution analysis of DNA copy number variation using comparative genomic hybridization to microarrays. Nat Genet (1998) 20(2):207–11. doi:10.1038/2524

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Veltman IM, Veltman JA, Arkesteijn G, Janssen IM, Vissers LE, de Jong PJ, et al. Chromosomal breakpoint mapping by arrayCGH using flow-sorted chromosomes. Biotechniques (2003) 35(5):1066–70.

PubMed Abstract | Google Scholar

5. Fiegler H, Gribble SM, Burford DC, Carr P, Prigmore E, Porter KM, et al. Array painting: a method for the rapid analysis of aberrant chromosomes using DNA microarrays. J Med Genet (2003) 40(9):664–70. doi:10.1136/jmg.40.9.664

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Kalscheuer VM, FitzPatrick D, Tommerup N, Bugge M, Niebuhr E, Neumann LM, et al. Mutations in autism susceptibility candidate 2 (AUTS2) in patients with mental retardation. Hum Genet (2007) 121(3–4):501–9. doi:10.1007/s00439-006-0284-0

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Abel HJ, Duncavage EJ. Detection of structural DNA variation from next generation sequencing data: a review of informatic approaches. Cancer Genet (2013) 206(12):432–40. doi:10.1016/j.cancergen.2013.11.002

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Chen W, Kalscheuer V, Tzschach A, Menzel C, Ullmann R, Schulz MH, et al. Mapping translocation breakpoints by next-generation sequencing. Genome Res (2008) 18(7):1143–9. doi:10.1101/gr.076166.108

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Chen W, Ullmann R, Langnick C, Menzel C, Wotschofsky Z, Hu H, et al. Breakpoint analysis of balanced chromosome rearrangements by next-generation paired-end sequencing. Eur J Hum Genet (2010) 18(5):539–43. doi:10.1038/ejhg.2009.211

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Harewood L, Kishore K, Eldridge MD, Wingett S, Pearson D, Schoenfelder S, et al. Hi-C as a tool for precise detection and characterisation of chromosomal rearrangements and copy number variation in human tumours. Genome Biol (2017) 18(1):125. doi:10.1186/s13059-017-1253-8

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Paterson AL, Weaver JM, Eldridge MD, Tavare S, Fitzgerald RC, Edwards PA, et al. Mobile element insertions are frequent in oesophageal adenocarcinomas and can mislead paired-end sequencing analysis. BMC Genomics (2015) 16:473. doi:10.1186/s12864-015-1685-z

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Dekker J, Marti-Renom MA, Mirny LA. Exploring the three-dimensional organization of genomes: interpreting chromatin interaction data. Nat Rev Genet (2013) 14(6):390–403. doi:10.1038/nrg3454

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Lieberman-Aiden E, van Berkum NL, Williams L, Imakaev M, Ragoczy T, Telling A, et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science (2009) 326(5950):289–93. doi:10.1126/science.1181369

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Belton J-M, McCord RP, Gibcus J, Naumova N, Zhan Y, Dekker J. Hi-C: a comprehensive technique to capture the conformation of genomes. Methods (2012) 58(3):268–76. doi:10.1016/j.ymeth.2012.05.001

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Burton JN, Adey A, Patwardhan RP, Qiu R, Kitzman JO, Shendure J. Chromosome-scale scaffolding of de novo genome assemblies based on chromatin interactions. Nat Biotechnol (2013) 31(12):1119–25. doi:10.1038/nbt.2727

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Engreitz JM, Agarwala V, Mirny LA. Three-dimensional genome architecture influences partner selection for chromosomal translocations in human disease. PLoS One (2012) 7(9):e44196. doi:10.1371/journal.pone.0044196

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Barutcu AR, Lajoie BR, McCord RP, Tye CE, Hong D, Messier TL, et al. Chromatin interaction analysis reveals changes in small chromosome and telomere clustering between epithelial and breast cancer cells. Genome Biol (2015) 16:214. doi:10.1186/s13059-015-0768-0

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Zhang CZ, Leibowitz ML, Pellman D. Chromothripsis and beyond: rapid genome evolution from complex chromosomal rearrangements. Genes Dev (2013) 27(23):2513–30. doi:10.1101/gad.229559.113

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Stephens PJ, Greenman CD, Fu B, Yang F, Bignell GR, Mudie LJ, et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell (2011) 144(1):27–40. doi:10.1016/j.cell.2010.11.055

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Zhang CZ, Spektor A, Cornils H, Francis JM, Jackson EK, Liu S, et al. Chromothripsis from DNA damage in micronuclei. Nature (2015) 522(7555):179–84. doi:10.1038/nature14493

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Baca SC, Prandi D, Lawrence MS, Mosquera JM, Romanel A, Drier Y, et al. Punctuated evolution of prostate cancer genomes. Cell (2013) 153(3):666–77. doi:10.1016/j.cell.2013.03.021

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Bagherani N, Smoller BR. An overview of cutaneous T cell lymphomas. F1000Res (2016) 5. doi:10.12688/f1000research.8829.1

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Elenitoba-Johnson KS, Wilcox R. A new molecular paradigm in mycosis fungoides and Sezary syndrome. Semin Diagn Pathol (2017) 34(1):15–21. doi:10.1053/j.semdp.2016.11.002

CrossRef Full Text | Google Scholar

24. Kaltoft K, Bisballe S, Rasmussen HF, Thestrup-Pedersen K, Thomsen K, Sterry W. A continuous T-cell line from a patient with Sézary syndrome. Arch Dermatol Res (1987) 279(5):293–8. doi:10.1007/BF00431220

CrossRef Full Text | Google Scholar

25. Heinz S, Benner C, Spann N, Bertolino E, Lin YC, Laslo P, et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell (2010) 38(4):576–89. doi:10.1016/j.molcel.2010.05.004

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Saldanha AJ. Java Treeview – extensible visualization of microarray data. Bioinformatics (2004) 20(17):3246–8. doi:10.1093/bioinformatics/bth349

CrossRef Full Text | Google Scholar

27. Rao SS, Huntley MH, Durand NC, Stamenova EK, Bochkov ID, Robinson JT, et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell (2014) 159(7):1665–80. doi:10.1016/j.cell.2014.11.021

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Krzywinski M, Schein J, Birol I, Connors J, Gascoyne R, Horsman D, et al. Circos: an information aesthetic for comparative genomics. Genome Res (2009) 19(9):1639–45. doi:10.1101/gr.092759.109

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Teng L, He B, Wang J, Tan K. 4DGenome: a comprehensive database of chromatin interactions. Bioinformatics (2015) 31(15):2560–4. doi:10.1093/bioinformatics/btv158

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Steininger A, Mobs M, Ullmann R, Kochert K, Kreher S, Lamprecht B, et al. Genomic loss of the putative tumor suppressor gene E2A in human lymphoma. J Exp Med (2011) 208(8):1585–93. doi:10.1084/jem.20101785

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Wickham H. Reshaping data with the reshape package. J Stat Softw (2007) 21(12):1–20. doi:10.18637/jss.v021.i12

CrossRef Full Text | Google Scholar

32. Wickham H. ggplot2: Elegant Graphics for Data Analysis. New York: Springer-Verlag (2009).

Google Scholar

33. Izykowska K, Zawada M, Nowicka K, Grabarczyk P, Braun FCM, Delin M, et al. Identification of multiple complex rearrangements associated with deletions in the 6q23–27 region in Sezary syndrome. J Invest Dermatol (2013) 133(11):2617–25. doi:10.1038/jid.2013.188

CrossRef Full Text | Google Scholar

34. Izykowska K, Przybylski GK, Gand C, Braun FC, Grabarczyk P, Kuss AW, et al. Genetic rearrangements result in altered gene expression and novel fusion transcripts in Sezary syndrome. Oncotarget (2017) 8(24):39627–39. doi:10.18632/oncotarget.17383

CrossRef Full Text | Google Scholar

35. Chen K, Wallis JW, McLellan MD, Larson DE, Kalicki JM, Pohl CS, et al. BreakDancer: an algorithm for high-resolution mapping of genomic structural variation. Nat Methods (2009) 6(9):677–81. doi:10.1038/nmeth.1363

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Afgan E, Baker D, van den Beek M, Blankenberg D, Bouvier D, Cech M, et al. The Galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2016 update. Nucleic Acids Res (2016) 44(W1):W3–10. doi:10.1093/nar/gkw343

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Rice P, Longden I, Bleasby A. EMBOSS: the European molecular biology open software suite. Trends Genet (2000) 16(6):276–7. doi:10.1016/S0168-9525(00)02024-2

CrossRef Full Text | Google Scholar

38. Karolchik D, Hinrichs AS, Furey TS, Roskin KM, Sugnet CW, Haussler D, et al. The UCSC table browser data retrieval tool. Nucleic Acids Res (2004) 32(Database issue):D493–6. doi:10.1093/nar/gkh103

CrossRef Full Text | Google Scholar

39. Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics (2010) 26(6):841–2. doi:10.1093/bioinformatics/btq033

PubMed Abstract | CrossRef Full Text | Google Scholar

40. O’Leary NA, Wright MW, Brister JR, Ciufo S, Haddad D, McVeigh R, et al. Reference sequence (RefSeq) database at NCBI: current status, taxonomic expansion, and functional annotation. Nucleic Acids Res (2016) 44(D1):D733–45. doi:10.1093/nar/gkv1189

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Phipson B, Smyth GK. Permutation P-values should never be zero: calculating exact P-values when permutations are randomly drawn. Stat Appl Genet Mol Biol (2010) 9:Article39. doi:10.2202/1544-6115.1585

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Lajoie BR, Dekker J, Kaplan N. The Hitchhiker’s guide to Hi-C analysis: practical guidelines. Methods (2015) 72:65–75. doi:10.1016/j.ymeth.2014.10.031

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Ramani V, Cusanovich DA, Hause RJ, Ma W, Qiu R, Deng X, et al. Mapping 3D genome architecture through in situ DNase Hi-C. Nat Protoc (2016) 11(11):2104–21. doi:10.1038/nprot.2016.126

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Chakraborty A, Ay F. Identification of copy number variations and translocations in cancer cells from Hi-C data. Bioinformatics (2018) 34(2):338–45. doi:10.1093/bioinformatics/btx664

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Edge P, Bafna V, Bansal V. HapCUT2: robust and accurate haplotype assembly for diverse sequencing technologies. Genome Res (2017) 27(5):801–12. doi:10.1101/gr.213462.116

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Selvaraj S, Dixon JR, Bansal V, Ren B. Whole-genome haplotype reconstruction using proximity-ligation and shotgun sequencing. Nat Biotechnol (2013) 31(12):1111–8. doi:10.1038/nbt.2728

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Dzamba M, Ramani AK, Buczkowicz P, Jiang Y, Yu M, Hawkins C, et al. Identification of complex genomic rearrangements in cancers using CouGaR. Genome Res (2017) 27(1):107–17. doi:10.1101/gr.211201.116

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Mansfield AS, Murphy SJ, Harris FR, Robinson SI, Marks RS, Johnson SH, et al. Chromoplectic TPM3-ALK rearrangement in a patient with inflammatory myofibroblastic tumor who responded to ceritinib after progression on crizotinib. Ann Oncol (2016) 27(11):2111–7. doi:10.1093/annonc/mdw405

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Choi J, Goh G, Walradt T, Hong BS, Bunick CG, Chen K, et al. Genomic landscape of cutaneous T cell lymphoma. Nat Genet (2015) 47(9):1011–9. doi:10.1038/ng.3356

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Kim N, Jinks-Robertson S. Transcription as a source of genome instability. Nat Rev Genet (2012) 13(3):204–14. doi:10.1038/nrg3152

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Meaburn KJ, Misteli T, Soutoglou E. Spatial genome organization in the formation of chromosomal translocations. Semin Cancer Biol (2007) 17(1):80–90. doi:10.1016/j.semcancer.2006.10.008

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Zhang Y, McCord RP, Ho Y-J, Lajoie BR, Hildebrand DG, Simon AC, et al. Chromosomal translocations are guided by the spatial organization of the genome. Cell (2012) 148(5):908–21. doi:10.1016/j.cell.2012.02.002

CrossRef Full Text | Google Scholar

53. Mathas S, Kreher S, Meaburn KJ, Johrens K, Lamprecht B, Assaf C, et al. Gene deregulation and spatial genome reorganization near breakpoints prior to formation of translocations in anaplastic large cell lymphoma. Proc Natl Acad Sci U S A (2009) 106(14):5831–6. doi:10.1073/pnas.0900912106

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Iborra FJ, Pombo A, Jackson DA, Cook PR. Active RNA polymerases are localized within discrete transcription ‘factories’ in human nuclei. J Cell Sci (1996) 109(Pt 6):1427–36.

Google Scholar

55. Osborne CS. Molecular pathways: transcription factories and chromosomal translocations. Clin Cancer Res (2014) 20(2):296–300. doi:10.1158/1078-0432.CCR-12-3667

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: chromosome conformation capture, chromoplexy, chromosomal translocations, deep sequencing, cutaneous T-cell lymphoma

Citation: Steininger A, Ebert G, Becker BV, Assaf C, Möbs M, Schmidt CA, Grabarczyk P, Jensen LR, Przybylski GK, Port M, Kuss AW and Ullmann R (2018) Genome-Wide Analysis of Interchromosomal Interaction Probabilities Reveals Chained Translocations and Overrepresentation of Translocation Breakpoints in Genes in a Cutaneous T-Cell Lymphoma Cell Line. Front. Oncol. 8:183. doi: 10.3389/fonc.2018.00183

Received: 14 November 2017; Accepted: 09 May 2018;
Published: 30 May 2018

Edited by:

Michael Breitenbach, University of Salzburg, Austria

Reviewed by:

Luisa Lanfrancone, Istituto Europeo di Oncologia s.r.l., Italy
Wei Chen, Southern University of Science and Technology, China

Copyright: © 2018 Steininger, Ebert, Becker, Assaf, Möbs, Schmidt, Grabarczyk, Jensen, Przybylski, Port, Kuss and Ullmann. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Reinhard Ullmann, reinhard1ullmann@bundeswehr.org

Present address: Anne Steininger, Steglitz, Berlin, Germany;
Grit Ebert, TMF – Technology, Methods, and Infrastructure for Networked Medical Research, Berlin, Germany

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.