Next Article in Journal
Mechanisms that Link Chronological Aging to Cellular Quiescence in Budding Yeast
Next Article in Special Issue
Integrated Analysis of Tissue-Specific Promoter Methylation and Gene Expression Profile in Complex Diseases
Previous Article in Journal
Insulin Resistance Does Not Impair Mechanical Overload-Stimulated Glucose Uptake, but Does Alter the Metabolic Fate of Glucose in Mouse Muscle
Previous Article in Special Issue
Familial Infertility (Azoospermia and Cryptozoospermia) in Two Brothers—Carriers of t(1;7) Complex Chromosomal Rearrangement (CCR):  Molecular Cytogenetic Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Molecular Cytogenomic Characterization of the Murine Breast Cancer Cell Lines C-127I, EMT6/P and TA3 Hauschka

1
Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Am Klinikum 1, D-07747 Jena, Germany
2
Croatian Institute for Brain Research, School of Medicine University of Zagreb, Salata 12, 10000 Zagreb, Croatia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(13), 4716; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21134716
Submission received: 16 June 2020 / Revised: 26 June 2020 / Accepted: 1 July 2020 / Published: 1 July 2020
(This article belongs to the Collection Feature Papers in Molecular Genetics and Genomics)

Abstract

:
Background: To test and introduce effective and less toxic breast cancer (BC) treatment strategies, animal models, including murine BC cell lines, are considered as perfect platforms. Strikingly, the knowledge on the genetic background of applied BC cell lines is often sparse though urgently necessary for their targeted and really justified application. Methods: In this study, we performed the first molecular cytogenetic characterization for three murine BC cell lines C-127I, EMT6/P and TA3 Hauschka. Besides fluorescence in situ hybridization-banding, array comparative genomic hybridization was also applied. Thus, overall, an in silico translation for the detected imbalances and chromosomal break events in the murine cell lines to the corresponding homologous imbalances in humans could be provided. The latter enabled a comparison of the murine cell line with human BC cytogenomics. Results: All three BC cell lines showed a rearranged karyotype at different stages of complexity, which can be interpreted carefully as reflectance of more or less advanced tumor stages. Conclusions: Accordingly, the C-127I cell line would represent the late stage BC while the cell lines EMT6/P and TA3 Hauschka would be models for the premalignant or early BC stage and an early or benign BC, respectively. With this cytogenomic information provided, these cell lines now can be applied really adequately in future research studies.

1. Introduction

Breast cancer (BC) is among the most common female specific cancer types and the second most common cancer in humans after lung cancer [1,2]. Survival rates of BC patients increased in the last years, especially in countries with early diagnostic regimens [3].
There are five stages of BC: (i) benign, premalignant stage; (ii) atypical ductal hyperplasia; (ii) preinvasive stage of ductal carcinoma in situ; (vi) metastatic carcinoma; and (v) advanced stage [4]. Known genetic changes in BC include acquired but also inherited changes in oncogenes, tumor suppressor genes and/or genes responsible for genomic stability [5,6]. Nowadays, such changes can be used as biomarkers for BC progression [7]. Moreover, BC can also be grouped according to immunohistochemical markers (ICM), like (i) presence or absence of receptors like those for estrogen (ER), progesterone (PR), human epidermal growth factor receptor-2 (HER-2) or epidermal growth factor receptor (EGFR) on tumor cell surface; (ii) expression of nuclear protein Ki67 as a marker of cell proliferation; and (iii) cytokeratin 5 expression in the plasma of BC cells. These and maybe more ICMs, like TP53 or androgen receptor gene expression, lead to subgrouping of BC in luminal A and B, HER2 positive and negative, and triple-negative or basal like subtypes [8,9,10] (Table 1).
BC subtyping is extremely important for metastasis staging and treatment [7,10,12,13]. In the majority of BC cases, surgical excision of primary tumor is the initial treatment step. Afterwards, radio- and especially chemotherapeutic options are legion; accordingly, various “Clinical Decision Support Systems” are available based on which the potentially most advantageous treatment options for individual patients may be found [14]. Important to mention here is also that gene mutations and specific acquired molecular signatures recently had some impact on the advance of therapeutic targets in breast cancer treatment [15,16].
However, the need for new types of medication with less side effects and being best targeted is still high [3]. Therefore, animal, especially murine, models are regarded as a highly feasible way not only to study biological pathways involved in initiation, progression and metastasis of a tumor like BC but also to establish new targeted medication, like murine tumor cell lines [16,17,18,19]. In spite of the widespread use of such murine tumor cell lines in research, surprisingly, most of them are not characterized molecular cytogenomically [20].
Fluorescence in situ hybridization (FISH) is considered the most practicable technique to detect gross genetic alteration in cancer [2]. Thus, in this study, it was taken advantage from combining multicolor-FISH using whole chromosome painting (wcp) probes, FISH banding [21], i.e., murine multicolor banding (mcb), and array-comparative genomic hybridization (aCGH) to do a first cytogenomic characterization of the three murine BC cell lines C-127I, EMT6/P and TA3 Hauschka, which is more than timely, as these cell lines were already established in 1978 [22], in 1986 [23] and in 1953 [24], respectively. Cell lines C-127I and EMT6/P were induced in mice by Harvey virus [22] and anthracycline treatment [23], respectively, while TA3 Hauschka was derived from tumorigenic ascites of a natural murine BC [24].
As previously done in comparable studies in murine tumor cell lines, a successful in silico translation from murine to human genome determined the corresponding homologous genetic alteration in human BC and enabled a classification as murine late stage, premalignant stage and benign BC-models.

2. Results

2.1. FISH Results

2.1.1. C-127I

Murine BC cell line C-127I presented as a pentaploid but was basically genetically relatively instable and was rearranged with more than 10 derivative chromosomes. Consequently, this cell line can be divided to four clones, clone 1 being the ancestor clone and clones 2, 3 and 4 being derivatives of that.
Clone 1 was present in 20% of the cells and can be described as 94~96,XXX,-2,der(4)t(4;10)(C4;C1),der(4)t(4;10)(C4;C1),+der(4)t(4;5)(C4;F),der(6)(pter→A1::G3→E::B3→D:),del(6)(D),+del(6)(A2),−7,+8,−9,dic(11;18)(A1;A1),inv(12)(BE),−12,−12,del(13)(B1),del(14)(D3),−14,+15,−16,+17,−18,−18,+19.
Clone 2, being present in 40% of the cells had the same karyotype as clone 1 with two additional aberrations, i.e., loss of one chromosome 8 and +der(17)t(11;17)(A3;B3) instead of a normal chromosome 17, as shown in Figure 1.
For clone 3, representing 23% of the cells compared to clone 1, one chromosome 12 was lost and one was replaced by a derivative chromosome 12: del(12)(A1.1).
Clone 4 formed the remaining 17% of cells—here, an additional der(5)t(5;13) and a idic(18) were present as a structural aberrations and one chromosome 10 and 18 each were lost.

2.1.2. EMT6/P

This cell line EMT6/P is triploid and can be divided into five main clones which show some chromosomal instability. Clone 2 can possibly be considered the “ancestor” clone.
The largest clone (clone 1) represents 40% of the cells, with the following karyotype (Figure 2): 59~64,X,der(X)t(X;5)(Xpter→XA1::XA6→XF5::5C3→5qter),der(3)(pter→H1::H1→F2:),+der(3)(pter→H1::H1→F2:),der(4)(pter→C5::E2→C5::C5→D2:),der(5)(5A1→5C3::5B1→5C3::15D1→15qter),+6,idic(8)(A1;A1),+8,idic(10)(A1;A1),idic(12)(A1;A1),dup(13)(C3A2),idic(14)(A1;A1),dup(15)(CA2),+17,+19.
In clone 2 (20%), the breakpoints of der(5)t(5;15) were different than in clone 1 — here, they were in 5B1 and 15D1 as der(5)t(5;15)(B1;D1) compared to a der(5)(5A1→5C3::5B1→5C3::15D1→15qter) in clone 1.
Clone 3 (10%) compared to clone 1 had no normal chromosome 12 and instead a second idic(12)(A1A1).
Clone 4 (7%) had no additional chromosome 3 and a complex reciprocal translocation t(2;3) with a der(3)t(2;3)(3pter→3H1::2?→2?qter) and der(2)t(2;3)(2?pter→2?::3H1→3F2:): the clone could not be found in mcb2 analyses; thus, breakpoints for chromosome 2 could not be determined.
Clone 5 (23%) showed reciprocal translocations as follows: t(2;11)(C3;D) and t(9;18); moreover, +der (9) t(9;18)(9pter→9?::18A2→18qter) and +der(18)t(9;18)(18pter→18E2::9?→9qter) were present compared to clone 1.

2.1.3. TA3 Hauschka

This cell line TA3 Hauschka had a particularly stable diploid karyotype and can be divided in three clones for which probably clone 1 was an ancestor of clones 2 and 3.
Kayrotype of clone 1 is shown in Figure 3 and can be described as follows:
41,X,der(X)(XA1→XA5::19C2→19D2::19D1→19qter),t(1;7)(B;F2),t(1;14)(G;E1),inv(3)(E1H4),del(4)(C3D2),t(6;16)(F3;C3),+6,t(12;13)(E;C1),+16,der(19)t(X;19)(C2;A6),del(19)(D2).
Clone 2 (30%) was characterized by an additional reciprocal translocation t(3;11)(G;E1) and a more complex derivative of chromosome 19, der(19)(19pter→19C2::XA6→XF5::8D1→8qter), compared to clone 1.
Clone 3 (20%) differed from clone 1 by a der(4)t(4;6;16)(A2;F3;C3) instead of del(4)(C3D2).

2.2. aCGH Results

The aforementioned FISH studies of the three murine BC cell lines were in line with the aCGH results and are summarized in Figure 4a, Figure 5a and Figure 6a. A translation of those results to the human genome (only imbalances larger than 3.5 megabase pairs were included in the evaluation) identified the corresponding homologous region in the human genome (Figure 4b, Figure 5b and Figure 6b). Genomic details can be found in Supplementary Table S1.

2.3. Comparison with Literature

The three studied BC cell lines present acquired copy number variations in regions known to harbor oncogenes and tumor suppressor genes, related to human BC [25,26]; as summarized in Table 2, gains of copy numbers were more frequent than losses. In Table 3, the chromosomal breakpoints observed in the three cell lines are compared to the break events known from human BC; here again, the highest rate of breaks being in concordance with human BC is present for the most advanced cell lines C-127I. Specific DNA copy number alterations correlated with the molecular subtype for human BC [27]—a comparison of the three murine BC cell lines is shown in Table 4. As a result, a high correspondence between C-127I and BC subtype HER2+ and basal-like tumors but also luminal B type was visible.

3. Discussion

BC has several subtypes (Table 1) due to the heterogeneity and complex pathology [13]. Still, animal model systems play a major role in BC research, especially for testing new treatment protocols [4,13]. Thus, in this study, the three frequently used murine BC cell lines C-127I, EMT6/P and TA3 Hauschka were for the first time characterized on the molecular cytogenomic level. The feasibility of the applied scheme was shown in several previous studies [20,36,37,38,39].
The three cell lines showed differences in the genomic alteration rate regarding ploidy, numerical and structural aberrations, and tumor-associated breakpoints. C-127I presented a very complex karyotype with pentaploidy, EMT6/P just was triploid and had less structural aberrations than C-127I, while TA3-Hauschka was near diploid and had only few imbalances. Thus, this is the first evidence that the three cell lines may demonstrate different subtypes and stages of human BC [40]. However, polyploidization is common in cancer cell lines [41,42], but polyploidization may also be part of malignancy progression and was also observed in connection with drug resistance [42]. Therefore, the pentaploidy in cell line C-127I may indicate that this cell line may be a suited model for the aggressive stage of BC or the HER2-enriched subtype which is known as an aggressive subtype and resistant to treatment [11,34].
As all three cell lines were established between 34 and 67 years ago [22,23,24], the chromosomal changes may be both original tumor related or acquired during long times of in vitro cultivation [43,44,45]. Only for TA3 Hauschka, the chromosome number at establishment is known, i.e., 41; thus, this cell line was, as other murine tumor cell lines, remarkably stable over times [24].
As highlighted in Table 2, CNVs could be observed in the three studied cell lines for 7, 10 or 17 of 21 regions known to be locations of tumor suppressor and/or oncogenes in human BC. The most aberrant was again C-127I, followed by EMT6/P and TA3 Hauschka. Interestingly, all three cell lines had gains of copy numbers in the region where the murine erbb2 gene is localized, while gain in the brca1 region was only evident for C-127I. Also, oncogene myc, being amplified in many human tumors, showed gain of copy numbers in C-127I and EMT6/P but not in TA3 Hauschka. Tumor suppressor gene rb1 was only deleted in C-127I.
As according to ploidy, karyotype and CNVs, C-127I is the most aberrant cell line, followed by EMT6/P and TA3 Hauschka; the same tendency is emphasized when looking at the number of chromosomal breakpoints and their localizations (Table 3). The murine chromosomal band homologous to human 9p21, which harbors tumor suppressor genes important in retinoblastoma and p53 pathway regulation, is affected by chromosomal break events in all three studied murine cell lines. The aforementioned pathways are affected in aggressive forms of BC [28,46]. The murine homologous band to human 19p13.1 is involved also in break events in all three studied cell lines, and this region was identified to comprise genes correlated for enhanced BC risk [29,31,47]. Furthermore, mutations in this region are strongly associated only with ER-negative BC subtypes [33]. Moreover, in TA3 Hauschka and C-127I, breakpoints homologous to human band 14q32 could be observed. Notably, there, the gene DICER1 is located, recently identified as playing a role in cancer predisposition [30,32].
Horlings and coworkers correlated in 2010 [27] the frequency for gains and losses of CNVs to BC molecular subtypes. For this study, only C-127I could be clearly assigned and not only to one but to even three BC subtypes (Table 4): HER2+, basal-like tumors, and luminal B type.
Finally, one may speculate about the influence of breakpoints and/or CNVs of regions being involved in presenting ICM, like ER or PR; in none of the three cell lines, there are changes in regions being homologous to gene estrogen receptor 1 (ESR1; in human 6q25.1). However, there was a loss in gene estrogen receptor 2 (ESR2; in human 14q23.2~23.3) in C-127I. The latter cell line presents also gains in progesterone receptor gene (PR; in human 11q22.1).
In conclusion, the molecular cytogenomic study and in silico translation for the three here studied BC cell lines, C-127I, EMT6/P and TA3 Hauschka, revealed that they can be used as models for human BC at different stages of malignancy. TA3 Hauschka can be best considered as a model for benign BC, EMT6/P may be used to represent the premalignant or early malignant stage of BC, while C-127I can be the model for the advanced BC stage. These insights are important for future application of these cell lines in BC research and their adequate use.

4. Materials and Methods

4.1. Cell Lines

The fibroblast-like/adhesively growing cell lines C-127I (Catalogue number CLS 400134) was purchased from CLS Cell Lines Service GmbH (Eppelheim, Germany), while the EMT6/P- (Catalogue number ECACC 96042344) and TA3 Hauschka-cell line (Catalogue number ECACC 85061102) were obtained from European Collection of Authenticated Cell Cultures (Salisbury, UK). They were grown adherently in DMEM medium (C-127I) or in EMEM/EBSS medium (EMT6/P and TA3 Hauschka) according to company instructions. Cells were prepared cytogenetically as previously reported [20]; whole genomic DNA was extracted using the Blood & Cell Culture DNA Midi Kit (Qiagen, Hilden, Germany) [36]. Cell line-derived chromosome-preparations were subjected to molecular cytogenetic analysis and extracted DNA were subjected to aCGH analysis. Cells were harvested shortly before they reached confluency; other cell density tests were not undertaken.

4.2. Molecular Cytogenetics

FISH was performed as previously described [36]. In short, murine whole chromosome paints (“SkyPaintTM DNA Kit M-10 for Mouse Chromosomes”, Applied Spectral Imaging, Edingen-Neckarhausen, Germany) were used for multicolor-FISH (mFISH; results not shown), and murine chromosome-specific mcb probe mixes were used for FISH banding [21]. At least 30 metaphases were acquired and analyzed for each probe set using Zeiss Axioplan microscopy (Carl Zeiss Jena, Jena, Germany), equipped with ISIS software (MetaSystems, Altlussheim, Germany). aCGH was done according to standard procedures by “SurePrint G3 Mouse CGH Microarray, 4x180K” (Agilent Technologies, SantaClara, CA, USA) [36].

4.3. Data Analysis

The breakpoints and imbalances of the three studied BC cell lines were determined according to aCGH and mcb data and aligned to human homologous regions using Ensembl Browser, as previously described [37]. The obtained data was compared to genetic changes known from human BC according to literature mentioned.

Supplementary Materials

Author Contributions

T.L. developed the idea for the study; M.M. and S.A. did the FISH studies; M.R. performed the aCGH studies and did the preevaluation; M.M. and S.A. performed the overall data interpretation; T.L. and S.A. did the final paper drafting; all authors agreed on the final draft.

Funding

This research was funded by grant # 2013.032.1 of the Wilhelm Sander-Stiftung.

Acknowledgments

The technical support of Nadezda Kosyakova (Jena, Germany) is kindly acknowledged.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

Abbreviations according to ISCN (2016) [48] are not included in this list
aCGHarray comparative genomic hybridization
BCbreast cancer
CNVcopy number variant
EGFRepidermal growth factor receptor
ERestrogen receptor
FISHfluorescence in situ hybridization
HER-2human epidermal growth factor receptor-2
ICMimmunohistochemical markers
ISCNInternational System for Human Cytogenomic Nomenclature
mcbmurine multicolor banding
mFISHmulticolor-fluorescence in situ hybridization
PRprogesterone receptor
wcpwhole chromosome painting

References

  1. Karbownik, A.; Sobańska, K.; Płotek, W.; Grabowski, T.; Klupczynska, A.; Plewa, S.; Grześkowiak, E.; Szałek, E. The influence of the coadministration of the p-glycoprotein modulator elacridar on the pharmacokinetics of lapatinib and its distribution in the brain and cerebrospinal fluid. Invest. New Drugs 2020, 38, 574–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Okaly, G.V.; Panwar, D.; Lingappa, K.B.; Kumari, P.; Anand, A.; Kumar, P.; Chikkalingaiah, M.H.; Kumar, R.V. FISH and HER2/neu equivocal immunohistochemistry in breast carcinoma. Indian J. Cancer 2019, 56, 119–123. [Google Scholar] [CrossRef]
  3. Raihan, J.; Ahmad, U.; Yong, Y.K.; Eshak, Z.; Othman, F.; Ideris, A. Regression of solid breast tumours in mice by Newcastle disease virus is associated with production of apoptosis related-cytokines. BMC Cancer 2019, 19, 315. [Google Scholar] [CrossRef] [Green Version]
  4. Ye, Y.; Qiu, T.H.; Kavanaugh, C.; Green, J.E. Molecular mechanisms of breast cancer progression: Lessons from mouse mammary cancer models and gene expression profiling. Breast Dis. 2004, 19, 69–82. [Google Scholar] [CrossRef] [PubMed]
  5. Richardson, M.E.; Chong, H.; Mu, W.; Conner, B.R.; Hsuan, V.; Willett, S.; Lam, S.; Tsai, P.; Pesaran, T.; Chamberlin, A.C.; et al. DNA breakpoint assay reveals a majority of gross duplications occur in tandem reducing VUS classifications in breast cancer predisposition genes. Genet. Med. 2019, 21, 683–693. [Google Scholar] [CrossRef] [Green Version]
  6. Tang, M.E.; Varadan, V.; Kamalakaran, S.; Zhang, M.Q.; Dimitrova, N.; Hicks, J. Major chromosomal breakpoint intervals in breast cancer co-localize with differentially methylated regions. Front. Oncol. 2012. [Google Scholar] [CrossRef] [Green Version]
  7. Kikuchi-Koike, R.; Nagasaka, K.; Tsuda, H.; Ishii, Y.; Sakamoto, M.; Kikuchi, Y.; Fuku, S.; Miyagawa, Y.; Hiraike, H.; Kobayashi, T.; et al. Array comparative genomic hybridization analysis discloses chromosome copy number alterations as indicators of patient outcome in lymph node-negative breast cancer. BMC. Cancer. 2019, 19, 521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Dai, X.; Li, T.; Bai, Z.; Yang, Y.; Liu, X.; Zhan, J.; Shi, B. Breast cancer intrinsic subtype classification, clinical use and future trends. Am. J. Cancer Res. 2015, 5, 2929–2943. [Google Scholar]
  9. Tang, P.; Tse, G.M. Immunohistochemical surrogates for molecular classification of breast carcinoma: A 2015 update. Arch. Pathol. Lab. Med. 2016, 140, 806–814. [Google Scholar] [CrossRef] [Green Version]
  10. Kondov, B.; Milenkovikj, Z.; Kondov, G.; Peterusevska, G.; Baseska, N.; Tolevska, N.; Ivkovski, L.J. Retention of approximal guiding plane surfaces in removable partial skeletal prosthesis. Open Access Maced. J. Med. Sci. 2018, 6, 1120–1125. [Google Scholar] [CrossRef] [Green Version]
  11. Fragomeni, S.M.; Sciallis, A.; Jeruss, J.S. Molecular subtypes and local-regional control of breast cancer. Surg. Oncol. Clin. N. Am. 2018, 27, 95–120. [Google Scholar] [CrossRef] [PubMed]
  12. Chen, X.; Pei, Z.; Peng, H.; Zheng, Z. Exploring the molecular mechanism associated with breast cancer bone metastasis using bioinformatic analysis and microarray genetic interaction network. Medcine 2018, 97, e12032. [Google Scholar] [CrossRef] [PubMed]
  13. Yersal, O.; Barutca, S. Biological subtypes of breast cancer: Prognostic and therapeutic implications. World J. Clin. Oncol. 2014, 5, 412–424. [Google Scholar] [CrossRef]
  14. Mazo, C.; Kearns, C.; Mooney, C.; Gallagher, W.M. Clinical decision support systems in breast cancer: A systematic review. Cancers 2020, 12, 369. [Google Scholar] [CrossRef] [Green Version]
  15. Kalimutho, M.; Nones, K.; Srihari, S.; Duijf, P.H.G.; Waddell, N.; Khanna, K.K. Patterns of Genomic Instability in Breast Cancer. Trends. Pharm. Sci. 2019, 40, 198–211. [Google Scholar] [CrossRef] [PubMed]
  16. Lima, Z.S.; Ghadamzadeh, M.; Arashloo, F.T.; Amjad, G.; Ebadi, M.R.; Younesi, L. Recent advances of therapeutic targets based on the molecular signature in breast cancer: Genetic mutations and implications for current treatment paradigms. J. Hematol. Oncol. 2019, 12, 38. [Google Scholar] [CrossRef]
  17. Wronski, A.; Arendt, L.M.; Kuperwasser, C. Humanization of the mouse mammary gland. Methods. Mol. Bio. 2015, 1293, 173–186. [Google Scholar] [CrossRef]
  18. Jones, R.A.; Moorehead, R.A. Integrative analysis of copy number and gene expression data identifies potential oncogenic drivers that promote mammary tumor recurrence. Genes Chromosomes Cancer 2019, 58, 381–391. [Google Scholar] [CrossRef]
  19. Osborne, C.; Wilson, P.; Tripathy, D. Oncogenes and tumor suppressor genes in breast cancer: Potential diagnostic and therapeutic applications. Oncologist 2004, 9, 361–377. [Google Scholar] [CrossRef]
  20. Rhode, H.; Liehr, T.; Kosyakova, N.; Rincic, M.; Azawi, S.S.H. Molecular cytogenetic characterization of two murine colorectal cancer cell lines. OBM Genet. 2018, 2, 037. [Google Scholar] [CrossRef]
  21. Liehr, T.; Starke, H.; Heller, A.; Kosyakova, N.; Mrasek, K.; Gross, M.; Karst, C.; Glaser, M.; Fickelscher, I.; Kuechler, A.; et al. Multicolor fluorescence in situ hybridization (FISH) applied to FISH-banding. Cytogenet. Genome Res. 2006, 114, 240–244. [Google Scholar] [CrossRef] [PubMed]
  22. Lowy, D.R.; Rands, E.; Scolnick, E.M. Helper-independent transformation by unintegrated Harvey sarcoma virus DNA. J. Virol. 1978, 26, 291–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Twentyman, P.R.; Fox, N.E.; Wright, K.A.; Workman, P.; Broadhurst, M.J.; Martin, J.A.; Bleehen, N.M. The in vitro effects and cross-resistance patterns of some novel anthracyclines. Br. J. Cancer 1986, 53, 585–594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Hauschka, T.S. Cell population studies on mouse ascites tumors. Trans. N. Y. Acad. Sci. 1953, 16, 64–73. [Google Scholar] [CrossRef] [PubMed]
  25. Hall, J.M.; Zuppan, P.J.; Anderson, L.A.; Huey, B.; Carter, C.; King, M. Oncogenes and human breast cancer. Am. J. Hum. Genet. 1989, 44, 577–584. [Google Scholar]
  26. Oliveira, A.M.; Ross, J.S.; Fletcher, J.A. Tumor suppressor genes in breast cancer. Am. J. Clin. Pathol. 2005, 124, S16–S28. [Google Scholar] [CrossRef]
  27. Horlings, H.M.; Lai, C.; Nuyten, D.S.A.; Halfwerk, H.; Kristel, P.; van Beers, E.; Joosse, S.A.; Klijn, C.; Nederlof, P.M.; Reinders, M.J.T.; et al. Integration of DNA copy number alterations and prognostic gene expression signatures in breast cancer patients. Clin. Cancer Res. 2010, 16, 651–663. [Google Scholar] [CrossRef] [Green Version]
  28. Lebok, P.; Roming, M.; Kluth, M.; Koop, C.; Özden, C.; Taskin, B.; Hussein, K.; Lebeau, A.; Witzel, I.; Wölber, L.; et al. p16 overexpression and 9p21 deletion are linked to unfavorable tumor phenotype in breast cancer. Oncotarget 2016, 7, 81322–81331. [Google Scholar] [CrossRef] [Green Version]
  29. Lawrenson, K.; Kar, S.; McCue, K.; Kuchenbaeker, K.; Michailidou, K.; Tyrer, J.; Beesley, J.; Ramus, S.J.; Li, Q.; Delgado, M.K.; et al. Functional mechanisms underlying pleiotropic risk alleles at the 19p13.1 breast-ovarian cancer susceptibility locus. Nat. Commun. 2016, 7, 12675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Cheng, T.H.T.; Thompson, D.J.; O’Mara, T.A.; Painter, J.N.; Glubb, D.M.; Flach, S.; Lewis, A.; French, J.D.; Freeman-Mills, L.; Church, D.; et al. Five endometrial cancer risk loci identified through genome-wide association analysis. Nat. Genet. 2016, 48, 667–674. [Google Scholar] [CrossRef]
  31. Couch, F.J.; Gaudet, M.M.; Antoniou, A.C.; Ramus, S.J.; Kuchenbaecker, K.B.; Soucy, P.; Beesley, J.; Chen, X.; Wang, X.; Kirchhoff, T.; et al. Consortium of Investigators of Modifiers of BRCA1/2. Common variants at the 19p13.1 and ZNF365 loci are associated with ER subtypes of breast cancer and ovarian cancer risk in BRCA1 and BRCA2 mutation carriers. Cancer Epidemiol. Biomark Prev. 2012, 21, 645–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. de Kock, L.; Geoffrion, D.; Rivera, B.; Wagener, R.; Sabbaghian, N.; Bens, S.; Ellezam, B.; Soglio, D.B.; Ordóñez, J.; Sacharow, S.; et al. Multiple DICER1-related tumors in a child with a large interstitial 14q32 deletion. Genes Chromosomes Cancer 2018, 57, 223–230. [Google Scholar] [CrossRef] [PubMed]
  33. Lupicki, K.; Elifio-Espostio, S.; Fononseca, A.S.; Weber, S.H.; Sugita, B.; Langa, B.C.; Pereira, S.R.F.; Govender, D.; Panieri, E.; Hiss, D.C.; et al. Patterns of copy number alterations in primary breasttumors of South African patients and their impacton functional cellular pathways. Int. J. Oncol. 2018, 53, 2745–2757. [Google Scholar] [CrossRef] [Green Version]
  34. Antoniou, A.C.; Kuchenbaecker, K.B.; Soucy, P.; Beesley, J.; Chen, X.; McGuffog, L.; Lee, A.; Barrowdale, D.; Healey, S. Common variants at 12p11, 12q24, 9p21, 9q31.2 and in ZNF365 are associated with breast cancer risk for BRCA1 and/or BRCA2 mutation carriers. Breast Cancer Res. 2012, 14, R33. [Google Scholar] [CrossRef]
  35. Huret, J.-L.; Ahmad, M.; Arsaban, M.; Bernheim, A.; Cigna, J.; Desangles, F.; Guignard, J.C.; Jacquemot-Perbal, M.-C.; Labarussias, M.; Leberre, V.; et al. Atlas of genetics and cytogenetics in oncology and haematology in 2013. Nucleic. Acids. Res. 2013, 41, D920–D924. [Google Scholar] [CrossRef]
  36. Kubicova, E.; Trifonov, V.; Borovecki, F.; Liehr, T.; Rincic, M.; Kosyakova, N.; Hussein, S.S. First molecular cytogenetic characterization of murine malignant mesothelioma cell line AE17 and in silico translation to the human genome. Curr. Bioinform. 2017, 12, 11–18. [Google Scholar] [CrossRef]
  37. Leibiger, C.; Kosyakova, N.; Mkrtchyan, H.; Glei, M.; Trifonov, V.; Liehr, T. First molecular cytogenetic high resolution characterization of the NIH 3T3 cell line by murine multicolor banding. J. Histochem. Cytochem. 2013, 61, 306–312. [Google Scholar] [CrossRef] [Green Version]
  38. Steinacker, R.; Liehr, T.; Kosyakova, N.; Rincic, M.; Azawi, SS. Molecular cytogenetic characterization of two murine cancer cell lines derived from salivary gland. Biol. Commun. 2018, 63, 243–255. [Google Scholar] [CrossRef]
  39. Guja, K.; Liehr, T.; Rincic, M.; Kosyakova, N.; Hussein Azawi, S.S. Molecular cytogenetic characterization identified the murine B-cell lymphoma cell line A-20 as a model for sporadic Burkitt’s lymphoma. J. Histochem. Cytochem. 2017, 65, 669–677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Inic, Z.; Zegarac, M.; Inic, M.; Markovic, I.; Kozomara, Z.; Djurisic, I.; Inic, I.; Pupic, G.; Jancic, S. Difference between luminal and luminal B subtypes according to Ki-67, tumor size, and progesterone receptor negativity providing prognostic information. Clin. Med. Insights Oncol. 2014, 8, 107–111. [Google Scholar] [CrossRef] [Green Version]
  41. Levan, A.; Biesele, J.J. Role of chromosomes in cancerogenesis.; as studied in serial tissue culture of mammalian cells. Ann. N. Y. Acad Sci. 1958, 71, 1022–1053. [Google Scholar] [CrossRef]
  42. Tan, Z.; Chu, D.Z.V.; Chan, Y.J.A.; Lu, Y.E.; Rancati, G. Mammalian cells undergo endoreduplication in response to lactic acidosis. Sci. Rep. 2018, 8, 2890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Kasai, F.; Hirayama, N.; Ozawa, M.; Iemura, M.; Kohara, A. Changes of heterogeneous cell populations in the Ishikawa cell line during long-term culture: Proposal for an in vitro clonal evolution model of tumor cells. Genomics 2016, 107, 259–266. [Google Scholar] [CrossRef] [PubMed]
  44. Mc Granahan, N.; Swanton, C. Clonal heterogeneity and tumor evolution: Past, present, and the future. Cell 2017, 168, 613–628. [Google Scholar] [CrossRef]
  45. Hyman, E.; Kauraniemi, P.; Hautaniemi, S.; Wolf, M.; Mousses, S.; Rozenblum, E.; Ringnér, M.; Sauter, G.; Monni, O.; Elkahloun, A.; et al. Impact of DNA amplification on gene expression patterns in breast cancer. Cancer Res. 2002, 62, 6240–6245. [Google Scholar]
  46. Xie, H.; Rachakonda, P.S.; Heidenreich, B.; Nagore, E.; Sucker, A.; Hemminki, K.; Schadendorf, D.; Kumar, R. Mapping of deletion breakpoints at the CDKN2A locus in melanoma: Detection of MTAP-ANRIL fusion transcripts. Oncotarget 2016, 7, 16490–16504. [Google Scholar] [CrossRef] [Green Version]
  47. Liu, Y.; Walavalkar, N.M.; Dozmorov, M.G.; Rich, S.S.; Civelek, M.; Guertin, M.J. Identification of breast cancer associated variants that modulate transcription factor binding. PLoS Genet. 2017, 13, e1006761. [Google Scholar] [CrossRef]
  48. McGowan-Jordan, J.; Simons, A.; Schmid, M. (Eds.) International System for Human Cytogenomic Nomenclature 2016 (ISCN 2016); Karger: Basel, Switzerland; Unionville, MO, USA, 2016. [Google Scholar]
Figure 1. Murine multicolor banding (mcb) was applied on chromosomes of BC cell line C-127I: Typical pseudocolor banding for all 20 different murine chromosomes is shown for clone 2. This figure depicts the summary of 20 chromosome-specific fluorescence in situ hybridization (FISH)-experiments. Four translocations consisting of two different chromosomes each, are highlighted by frames in this summarizing karyogram. Chromosomes with partial deletions are pointed out by red arrows, and chromosomes with inversions are pointed out by blue arrows.
Figure 1. Murine multicolor banding (mcb) was applied on chromosomes of BC cell line C-127I: Typical pseudocolor banding for all 20 different murine chromosomes is shown for clone 2. This figure depicts the summary of 20 chromosome-specific fluorescence in situ hybridization (FISH)-experiments. Four translocations consisting of two different chromosomes each, are highlighted by frames in this summarizing karyogram. Chromosomes with partial deletions are pointed out by red arrows, and chromosomes with inversions are pointed out by blue arrows.
Ijms 21 04716 g001
Figure 2. Murine multicolor banding (mcb) was applied on chromosomes of BC cell line EMT6/P: Legend is as for Figure 1. Partial duplications are highlighted by green arrows.
Figure 2. Murine multicolor banding (mcb) was applied on chromosomes of BC cell line EMT6/P: Legend is as for Figure 1. Partial duplications are highlighted by green arrows.
Ijms 21 04716 g002
Figure 3. Murine multicolor banding (mcb) was applied on chromosomes of BC cell line TA4 Hauschka: Legend is as for Figure 1.
Figure 3. Murine multicolor banding (mcb) was applied on chromosomes of BC cell line TA4 Hauschka: Legend is as for Figure 1.
Ijms 21 04716 g003
Figure 4. (A) Array comparative genomic hybridization (aCGH) results for murine BC cell line C-127I: The copy number alterations with respect to the pentaploid karyotype are given as the color code depicted in the figure with shades of red (losses) and green (gains); purple arrows indicate breakpoints. Breakpoints are indicated according to mcb results. (B) Projection of the aCGH results for the cell line onto the human genome showing imbalances as gains and losses of specific chromosomal regions with respect to the original pentaploid chromosome set.
Figure 4. (A) Array comparative genomic hybridization (aCGH) results for murine BC cell line C-127I: The copy number alterations with respect to the pentaploid karyotype are given as the color code depicted in the figure with shades of red (losses) and green (gains); purple arrows indicate breakpoints. Breakpoints are indicated according to mcb results. (B) Projection of the aCGH results for the cell line onto the human genome showing imbalances as gains and losses of specific chromosomal regions with respect to the original pentaploid chromosome set.
Ijms 21 04716 g004
Figure 5. aCGH results for the triploid murine BC cell line EMT6/P (A) and its projection onto the human genome (B): For more details, see legend of Figure 4.
Figure 5. aCGH results for the triploid murine BC cell line EMT6/P (A) and its projection onto the human genome (B): For more details, see legend of Figure 4.
Ijms 21 04716 g005
Figure 6. aCGH results for near diploid murine BC cell line TA3 Hauschka (A) and its projection onto the human genome (B): For more details, see legend of Figure 4.
Figure 6. aCGH results for near diploid murine BC cell line TA3 Hauschka (A) and its projection onto the human genome (B): For more details, see legend of Figure 4.
Ijms 21 04716 g006
Table 1. Relationship between the molecular breast cancer (BC) subtypes and immunohistochemical markers (ICMs) [11].
Table 1. Relationship between the molecular breast cancer (BC) subtypes and immunohistochemical markers (ICMs) [11].
Molecular SubtypeERPRHER2
luminal A++
luminal B++
luminal B+++
HER-2++
triple negative or basal-like
ER—estrogen receptor; HER-2—human epidermal growth factor receptor 2; PR—progesterone receptor.
Table 2. Oncogenes and tumor suppresser genes of importance in BC according to the literature [25,26] and their involvement in gains or loss of copy numbers in the three studied cell lines. Abbreviation: CNV = copy number variant.
Table 2. Oncogenes and tumor suppresser genes of importance in BC according to the literature [25,26] and their involvement in gains or loss of copy numbers in the three studied cell lines. Abbreviation: CNV = copy number variant.
Oncogenes and Gene Loci in Human
Tumor Suppressor GenesC-127IEMT6/PTA3 Hauschka
NRAS1p22 or p13gaingainno CNV
MSH22p22gaingainno CNV
RAF13p25gaingaingain
RARβ23p24no CNVno CNVno CNV
MLH13p21lossno CNVno CNV
APC5q21gaingainno CNV
MYB6q22-q23gainno CNVno CNV
IGFII-R6q26gaingainno CNV
MYC8q24gaingainno CNV
CDKN2A (p16INK4)9p21lossgainloss
PTEN 10q2310q23gaingainno CNV
HRAS11p15.5lossno CNVno CNV
ATM11q22gainno CNVno CNV
CCND111q13gaingainno CNV
INT211q13lossgainno CNV
CDKN1B (p27kip1)12p13no CNVgaingain
KRAS212p12.1no CNVgaingain
BRCA213q12gainno CNVno CNV
RB113q14lossno CNVno CNV
CDH1 (E-cadherin)16q22no CNVno CNVgain
TP53 (p53)17p13gainno CNVno CNV
ERBB217q21gaingaingain
BRCA117q21gainno CNVno CNV
SERPINB5 (maspin)18q21lossno CNVno CNV
STK11 (LKB1)19p13gaingaingain
SUM of concordance in CNVs of potentially affected regions17/2110/217/21
Table 3. Breakpoints in C-127I, EMT6/P and TA3 Hauschka compared to the observed acquired breaks in human BCs according to the literature [6,7,11,25,26,28,29,30,31,32,33,34,35]: Concordances with human breakpoints are highlighted in bold.
Table 3. Breakpoints in C-127I, EMT6/P and TA3 Hauschka compared to the observed acquired breaks in human BCs according to the literature [6,7,11,25,26,28,29,30,31,32,33,34,35]: Concordances with human breakpoints are highlighted in bold.
Breakpoint
Acc. to Human Genome
Human BCC-127IEMT6/PTA3 Hauschka
1p33++
1p13.2+
1q25.3++
2p23.3+
2q31.3++
3p26.1++
3p12.3++
3q14.1+
3q21.3++
4p12+
4q22.3++
4q26++
4q31.23++
4q32.2+
5p14.2++
5q13.2++
5q14.3++
5q15+
6q12++
6q25.2+
7p14.1+
7q31.1+
7q36.2+
8q23.3++
8q24.22++
9p24.2++
9p21++++
10p11.21++
10q25.1+
11p15.5++
12p13.2+
12q12.1++
12q24.31++
13q21.2++
14q32+++
16p12.3+
16q13.3++
16q21+
17p12++
17q21++
19p13.1++++
20q13.3++
22q12.2++
Xp22.2+
Xq23.2+
SUM of concordance27/4519/4518/45
Table 4. Copy number changes associated with molecular subtypes of human BC, according to [27], with the copy number variants (CNVs) in cell lines C-127I, EMT6/P and TA3 Hauschka: Concordances with human CNVs (in italics) are highlighted in bold. Abbreviations: no CNV = no copy number variants.
Table 4. Copy number changes associated with molecular subtypes of human BC, according to [27], with the copy number variants (CNVs) in cell lines C-127I, EMT6/P and TA3 Hauschka: Concordances with human CNVs (in italics) are highlighted in bold. Abbreviations: no CNV = no copy number variants.
DNA Changes in BC SubtypesHuman BCC-127IEMT6/P TA4 Hauschka
HER2+
17q11.1~12gaingainno CNVno CNV
17q21.31~23.2gaingainno CNVno CNV
SUM of concordance2/20/20/2
Basal-like tumors
4p15.31lossno CNVgainno CNV
5q12.3~13.2lossno CNVno CNVno CNV
5q33.1losslossno CNVno CNV
6p12.3gaingaingainno CNV
6p21.1~23gaingaingainno CNV
8q24.21~24.22gaingaingainno CNV
10p12.33~14gainlossno CNVno CNV
10q23.33lossno CNVno CNVno CNV
12q13.13~13.3lossgaingainno CNV
15q15.1losslossno CNVno CNV
15q21.1losslossno CNVno CNV
SUM of concordance6/113/110/11
luminal A
1q21.3~44gainno CNVgainno CNV
16p13.12~13.13gainno CNVno CNVno CNV
16q11.2~13lossno CNVgaingain
16q22.1-24.1lossno CNVgainno CNV
SUM of concordance0/41/40/4
luminal B
1p31.3losslosslossloss
8p21.2~23.1lossno CNVgainno CNV
17q23.2gaingainno CNVno CNV
SUM of concordance2/31/31/3

Share and Cite

MDPI and ACS Style

Azawi, S.; Liehr, T.; Rincic, M.; Manferrari, M. Molecular Cytogenomic Characterization of the Murine Breast Cancer Cell Lines C-127I, EMT6/P and TA3 Hauschka. Int. J. Mol. Sci. 2020, 21, 4716. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21134716

AMA Style

Azawi S, Liehr T, Rincic M, Manferrari M. Molecular Cytogenomic Characterization of the Murine Breast Cancer Cell Lines C-127I, EMT6/P and TA3 Hauschka. International Journal of Molecular Sciences. 2020; 21(13):4716. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21134716

Chicago/Turabian Style

Azawi, Shaymaa, Thomas Liehr, Martina Rincic, and Mattia Manferrari. 2020. "Molecular Cytogenomic Characterization of the Murine Breast Cancer Cell Lines C-127I, EMT6/P and TA3 Hauschka" International Journal of Molecular Sciences 21, no. 13: 4716. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21134716

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop