Zum Hauptinhalt springen

Cytogenomic characterization of three murine malignant mesothelioma tumor cell lines

Wahlbuhl, Eva ; Liehr, Thomas ; et al.
In: Molecular Cytogenetics, Jg. 13 (2020), Heft 1, S. 1-10
Online academicJournal

Cytogenomic characterization of three murine malignant mesothelioma tumor cell lines 

Background: Malignant mesothelioma (MM) is a rare aggressive cancer primary located in pleura and lung. MMs can be divided into biphasic, epithelioid and sarcomatoid subtypes. In majority of cases MMs are induced by asbestos fiber exposure. As latency period after asbestos exposure ranges between ~ 10 and 60 years MMs are mainly observed in elder people. Human MM, being a rare tumor type, lacks detailed cytogenetic data, while molecular genetic studies have been undertaken more frequently. However, murine MM cell lines are also regularly applied to get more insight into MM biology and to test new therapy strategies. Results: Here the murine MM cell lines AB1, AB22 and AC29 were studied by molecular cytogenetics and molecular karyotyping. Interestingly, yet there were no genetic or genomic studies undertaken for these already in 1992 established cell lines. The obtained data on genomic imbalances in these murine cell lines was translated into the human genome as previously reported based on human and murine genomic browsers. Conclusions: It turned out that all three cell lines showed high similarities in copy number variants as observed typically in human MM. Also, all three cell lines were most similar to human epithelioid MMs, and should be used as models therefore.

Keywords: Murine multicolor banding (mcb); Array comparative genomic hybridization (aCGH); Malignant mesothelioma; Murine cell line; AB1; AB22; AC29

Electronic supplementary material The online version of this article (10.1186/s13039-020-00511-4) contains supplementary material, which is available to authorized users.

Background

Malignant mesothelioma (MM) is a rare aggressive tumor-family of pleura and lung, with an incidence of about 0.002% [[1]]. In most of the cases, MMs are located in pleural mesothelium, and only rarely in peritoneal cavities, tunica vaginalis or pericardium. MM can be specifically promoted by exposure to asbestos fibers [[3]]. Besides working with asbestos, accordingly contaminated buildings provide an additional, often unrecognized problem, where affected person can undergo asbestos inhalation, ingestion, or less often, severe exposures via the skin [[3], [5]]. The latency periods for MM after asbestos exposure can range from 1 to 6 decades, and the median age of onset is 72 years [[6]].

Numerous genetic changes are involved in MM. These include numerical and structural chromosomal aberrations and molecular genetically detectable alterations in the cellular signal transduction pathways, among others caused by activation of oncogenes or loss of tumor suppressor genes [[5]]. In human the genes cyclin-dependent kinase inhibitor 2A (CDKN2A), neurofibromatosis type 2 (NF2), the breast cancer associated gene 1 (BRCA1) associated protein 1 (BAP1) and tumorsuppressorprotein 53 (TP53) genes seem to be major players in MM-pathogenesis and -progression [[7]–[16]].

Histomorphologically and according to their growth parameters, MM can be divided into the following, most frequently observed subtypes: (1) biphasic, (2) epithelioid and (3) sarcomatoid. Different median survival times were attributed to each subtype; the best prognosis has the epithelioid, while the worst one has the sarcomatoid subtype [[13], [17]].

As MM is an aggressive tumor with poor prognosis, there is ongoing research to better understand the biology of this cancer type [[18]]. Therefore, also animal models, including murine tumor cell lines are regularly applied, also because human and mouse genomes show homologies within coding sequences of up to 97% [[20]]. In 1992 Davis and coworkers inoculated asbestos fibers into female BALB/c and CBA mice and established successfully 12 MM cell lines from tumor ascites cells [[21]]. Here two of these cell lines derived from BALB/c mice, i.e. AB1 and AB12, and one of them from CBA mice (AC29) were studied. Strikingly, in none of these cell lines (cyto)genetic research was undertaken yet to characterize their cytogenomic content. However, the latter data are important to use such cell lines in the best suited way to answer questions about MM-biology or to apply them in tests for new treatment options, i.e. for drug tests meant for the corresponding MM subtype.

Results

Molecular cytogenetics

Ab1

This cell line showed the following hypotetraploid composite karyotype (Fig. 1a) 73~80<4n>,-X,-X,der(1)t(1;2)(H5;F1),der(1)t(1;2)(H5;F1),+der(1)t(1;2)(C1;F1),-2,der(2)(2A1→2H4:6F3~G1→6E1:6F3~G1→6qter),der(2)(2A1→2H4:6F3~G1→6E1:6F3~G1→6qter),der(2)t(2;19)(E3;D1),+3,der(6)t(2;6)(H1;E1),der(6)t(2;6)(H1;E1),der(7)t(7;19)(E3;D1),dic(9;19)(A1;D3),dic(9;19)(A1;D3),del(13)(A5),dic(13;17)(A1;A1),dic(13;17)(A1;A1),der(15)t(15;?)(E1?;?),+der(15)(15pter→15E1:17B→17E3:17E3→17B:),+der(15)(15pter→15B2:17B→17E3:17E3→17B:),-16,-17,-17[11],-18[10],del(19)(D1),del(19)(D1).

Graph: Fig. 1 a Results of mFISH using all 21 murine whole chromosome paints as probes applied on murine MM cell line AB1 are shown here. b Typical pseudocolorbanding-results for murine multicolor banding (mcb) as applied on cell line AB1 for chromosomes 1 (mcb1), 2 (mcb2) and 6 (mcb6). Derivative chromosomes are shown as 1/2a = der(1)t(1;2)(H5;F1), 1/2b, = der(1)t(1;2)(C1;F1), 2/6 = der(2)(2A1→2H4:6F3~G1→6E1:6F3~G1→6qter) and 6/2 = der(6)t(2;6)(H1;E1); normal chromosomes are labeled by # and chromosome number

In Fig. 1b examples of mcb experiments are shown for chromosomes 1, 2 and 6, which enabled the characterization of the der(1)t(1;2)(C1;F1), the der(1)t(1;2)(H5;F1)x2 and the der(2)(2A1→2H4:6F3~G1→6E1:6F3~G1→6qter)x2.

Ab22

The tumor cell line AB22 was near tetraploid (Fig. 2a)—here the composite karyotype: 73~79<4n>,-X,-X,der(X)t(X;6)(C~D;C1),der(X)(XA1→XC~D:6C1→6G2:XF1→Xqter),dic(3;3)(A1;A1),del(3)(A3F1),der(4)(4A1→4C3:4C3:4C5→4C7:4C7→4C5:2F3→2qter),der(4)(4A1→4C3:4C3:4C5→4C7:4C7→4C5:2F3→2qter),-5,der(5)t(5;11)(G2;D~E),der(6)t(X;6)(D;C1),der(6)t(X;6)(D;C1),der(7)t(7;9)(F4;F1),der(7)t(7;9)(F4;F1),-10,-12,-13,-14,der(15)t(5;15)(G2;E),der(15)t(5;15)(G2;E),der(15)(15pter→15E:6D→6E:15E→15qter),+der(15)(15A1→15E:6D→6E:15E→15qter),+der(15)(15pter→15E:6D→6E:15E→15qter),del(16)(B2),der(16)(pter→B2:B1->qter),inv(17)(CE5),inv(17)(CE5),-18,+19.

Graph: Fig. 2 mFISH (a) and selected mcb results (b) for murine MM cell line AB22. For mcb3 two normal chromosomes (#3), a dic(3;3)(A1;A1) (dic(3;3)) and a del(3)(A3F1) (del(3)) are depicted. Also application of mcb17 revealed the presence of two normal chromosomes 17 (#17) and two chromosomes 17 with inversion inv(17)(CE5) (inv(17))

Here examples for the mcb characterization of dic(3;3)(A1;A1) and del(3)(A3F1) by mcb3 and of inv(17)(CE5)x2 are shown in Fig. 3b.

Graph: Fig. 3 a mFISH result of the main clone being present in 90% of the cell line AC29 is shown here. The only difference in the subclone comprising 10% of the cells is that the del(13)(A5B) is replaced by a der(13)t(13)t(12;13) as shown in white square. b Result for mcb9 and 15 highlight the dic(9;15)(A1;A1) here labeled as 9/15

Ac29

AC29 turned out to be hyper-triploid with one main clone (90%) and one smaller subclone (10%). The main clone karyotype (Fig. 3a) had the following karyotype: 63<3n>,X,-X,der(X)(pter→A1:A2→qter),der(1)(pter→D:E4→G:H2→qter),der(1)(pter→C5:C2→qter),del(2)(E2E5),der(2)(2pter→2H3:19C3→19D2:11D→11qter),+del(3)(A3E3),+del(3)(A3E3),+del(4)(C4),der(5)t(5;6)(B;B3),-6,der(6)t(6;12)(G1;C2),+der(8)(8pter→8A3:8B3→8E2:18D→18E4:1E4→1G:18D→18E4:1E4→1G:18E4→18D:1E4→1G),dic(9;15)(A1;A1),del(11)(B4E1),der(11)(pter→B4:A2→qter),der(11)t(2;11)(H3;D),der(12)t(6;12)(G1;C2),del(13)(A5B),der(17)t(11;17)(D;E5),der(17)(17pter→17B~C:6B1→6G3:6B1→6G3:17B~C→17E5:2H3→2qter),der(17)(17pter→17E5:17E4→17E5:11E1→11qter),der(18)t(5;18)(B1;D3),del(18)(B1C),+mar1,+mar2.

The subclone was just characterized by a translocation between chromosome 12 and 13 {der(13)t(13)t(12;13)} instead of del(13)(A5B) compared to the main clone (Fig. 3a).

As examples for mcb the characterization of the dic(9;15)(A1;A1) is shown in Fig. 3b.

Two marker chromosomes could be resolved here, neither by multicolor fluorescence in situ hybridization using all 21 murine whole chromosome paints as probes (mFISH—Fig. 3a) nor by mcb. Thus, most likely they are derivatives of the centromere-near region of any of the murine chromosomes—subband A1, which do not specifically stain by any euchromatic DNA-probe. Accordingly, the marker chromosomes could be left overs of the dic(9;15)(A1;A1) and a del(?)(A1).

aCGH

Array comparative genomic hybridization (aCGH) data (Additional file 1: Table 1) together with which FISH results could be summarized in Figs. 4a, 5a and 6a. These results were translated to the corresponding homologous regions in the human genome as depicted in Figs. 4b, 5b and 6b. All in the evaluation included imbalances were larger than 3.5 mega base pairs.

Graph: Fig. 4 aCGH results for cell line AB1. In a copy number variations detected are summarized with respect to a tetraploid karyotype. Gains are depicted as green bars (one more copy = light green; two more copies = dark green), loss of one copy is depicted as a red bar and loss of two copies is depicted as a dark-red bar. Breaks are registered here as arrows. In b results of in silico translation for AB1 to human genome are shown the same way as in a

Graph: Fig. 5 aCGH results for cell line AB22 depicted with respect to a tetraploid karyotype; legend like in Fig. 4

Graph: Fig. 6 aCGH results for cell line AC29 depicted with respect to a triploid karyotype; legend like in Fig. 4

Data-analyses

The common aberrations and cytogenetic changes that frequently occur in MM [[22]] revealed for all three cell lines to be less similar to human biphasic MM subtype (Table 1). According to Table 1, AB1 has 14/16 (88%) aberrations in common with human epithelioid and 17/21 (81%) aberrations with human sarcomatoid MMs. For AB22 it was 18/20 (94%) and 17/21 (81%) concordance to human epithelioid and sarcomatoid MMs, respectively. And for AC29 similarities of human epithelioid or sarcomatoid MMs was 15/20 (75%) versus 15/22 (68%). Thus, all three cell lines seemed to be best suited as models for human epithelioid MMs.

Comparison of 'translated' imbalances of murine MM-cellines AB1, AB22 and AC29 with human epitheloid, biphasic and sarcomatoid MM, according to Krismann et al. [[22]]

CNV detected in cell lines AB1

Epithelial MM

Biphasic MM

Sarcomatoid MM

del(1)(pter32)

?

+

(+)

amp(1)(p31q32)

+

(+)

del(2)(p23p16)

+

amp(2)(q12q21.2)

(+)

+

del(2)(q22q32)

?

del(3)(p22p10)

?

+

del(3)(q10q24)

?

amp(3)(q25q26)

?

?

+

del(3)(q27qter)

?

?

amp(5)(p15p12)

+

+

del(5)(p12q15)

?

?

+

amp(6)(p22.1q12)

(+)

(+)

(+)

del(6)(q22.3qter)

+

+

+

amp(8)(q11.2q21.2)

+

(+)

+

del(9)(pterqter)

+

+

+

del(10)(p15p12)

+

+

+

del(10)(q23q25)

(+)

(+)

(+)

del(11)(pterp10)

?

+

del(11)(p10qter)

?

+

+

amp(15)(q21.2q24)

+

+

?

del(18)(p11.2qter)

+

?

+

amp(20)(pterqter)

+

(+)

del(21)(q11.1qter)

?

?

del(X)(pterqter)

+

Sum for +

11/16

6/19

12/21

Sum for (+)

3/16

3/19

5/21

Sum for + and (+)

14/16

9/19

17/21

CNV detected in cell lines AB22

Epithelial MM

Biphasic MM

Sarcomatoid MM

del(1)(pter32)

+

+

(+)

amp(1)(p32p10)

+

+

del(3)(p24p24)

+

(+)

del(3)(p21.2p14.2)

?

?

(+)

amp(3)(p14.2q21)

+

+

+

del(3)(q26qter)

?

?

del(4)(pterq21.1)

+

+

+

amp(5)(p15.3p12)

+

+

del(5)(q11qter)

?

?

(+)

del(6)(q16q25)

+

+

+

amp(7)(pterp22)

+

+

?

del(7)(p22qter)

+

del(8)(p22p12)

+

+

+

amp(8)(q22qter)

+

(+)

+

del(9)(q21.2q22.3)

(+)

(+)

?

del(10)(pterq23.2)

+

+

+

amp(11)(q11q13.3)

?

?

del(12)(p12.1qter)

?

+

del(13)(q13q32)

+

+

+

del(14)(q11qter)

+

+

+

del(18)(pterqter)

+

(+)

+

del(19)(pterp13.3)

?

(+)

(+)

amp(20)(pterqter)

+

(+)

del(22)(q11.2q11.2)

+

+

+

del(22)(q13.2qter)

+

+

+

Sum for +

17/20

11/22

15/23

Sum for (+)

1/20

4/22

6/23

Sum for + and (+)

18/20

15/22

21/23

CNV detected in cell lines AC29

Epithelial MM

Biphasic MM

Sarcomatoid MM

amp(1)(p31 q25)

(+)

amp(2)(p23p11.2)

+

amp(2)(q33qter)

+

+

amp(3)(pterq24.3)

+

amp(3)(p14.1p13)

+

+

?

amp(3)(q25q26.2)

(+)

(+)

+

amp(5)(p15.2p13.1)

+

+

del(5)(q21q32)

?

?

(+)

amp(5)(q33qter)

?

+

del(6)(q15q16.1)

+

+

+

del(7)(p21p15.1)

+

+

?

amp(7)(p13p11.1)

del(7)(q21.3q36)

+

+

+

amp(8)(p12p11.1)

+

amp(8)(q22.2qter)

+

(+)

+

del(9)(pterq33)

+

+

+

del(13)(q13q14.1)

+

+

+

del(15)(q13q21.1)

(+)

del(17)(pterp12)

+

+

(+)

del(17)(q11q23)

?

amp(17)(q24qter)

+

(+)

amp(19)(p13.2p13.1)

?

amp(20)(q13.2qter)

+

+

del(X)(p22p11.2)

(+)

Sum for +

14/20

8/23

10/22

Sum for (+)

1/20

2/23

5/22

Sum for + and (+)

15/20

10/23

15/22

Only imbalances present in any of the three human MMs are listed CNV = copy number variation; + = aberration present in the cell line, − absent in the cell line; ? = no clear correlation possible, as it can be + or − in human cases

Also in Table 2 region, where four tumor suppressor genes meant to play important role in human MM are localized, were checked for copy number variant presence in the three studied murine MM cell lines. No correlations were found here.

The four tumor suppressor genes most often involved in human MM acc. to [[7]–[16]], being deleted and/or mutated there are compared for copy number variant observed in the three studied cell lines

Human gene/murine homologue

AB1

AB22

AC29

BAP1 deletion 3p21.1

(+)

+

No CNV

CDKN2A deletion 9p21.2

No CNV

No CNV

No CNV

TP53 deletion 17p13.1

No CNV

No CNV

No CNV

NF2 deletion 22q12.2

No CNV

No CNV

No CNV

Genomic locations (HG19/GRCh37): BAP1 - chr3:52,435,020-52,444,121; CDKN2A - chr9:21,967,751-21,994,490; TP53 - chr17:7,571,720-7,590,868; NF2 - chr22:29,999,545-30,094,589 + = partially deleted; (+) = possibly deleted; no CNV = no copy number alteration; dup = duplication instead of deletion

Conclusions

The murine MM cell lines AB1, AB22 and AC29 were studied in this paper for the first time by molecular cytogenetics combined with aCGH. This enabled to determine their genetic alterations and imbalances and align these with human MMs. mFISH using whole chromosome painting probes revealed the general characteristics of the cell lines, like the ploidy, clonal and nonclonal changes as well as numerical and intrachromosomal structural aberrations. By mcb interchromosomal alterations as duplications, deletions or inversions, and chromosomal breakpoints involved could be uncovered, as previously reported [[23]–[28]]. The aCGH data was aligned with the FISH results and also used to determine breakpoints of unbalanced rearrangements (Additional file 1: Table 1).

The cell lines A1 and AC29 were tetraploid; as in both cell lines derivative chromosomes were present twice, it is possible that polyploidization was a result of cell culture, and tetraploidy was absent in original tumor. Such so-called telomere-driven tetraploidization in the context of cell culture-related factors as trypsin treatment, increasing number of cell-culture passages, and oxygen exposure [[29]] was discussed before. However, as no karyotype of tumor or early cell passages of A1 and AC29 are available, this is just speculation and cannot be tested by any means.

Interestingly, a deletion of CDKN2A gene is considered as one of the most typical alterations in human MMs [[7]–[9]]. In the AB1 and AC29 there was indeed a deletion in the murine homologous region; however in cell line AB22 this region was duplicated (Additional file 1). For other tumor suppressor genes BAP1, NF2 and TP53 thought to play important roles in human MMs [[10]–[16]], there is even less or no concordance in the copy number variant regions of the three cell lines (Additional file 1).

Nonetheless, the overall similarities of copy number variants found in the three murine MM cell lines compared to human MM are striking. A shown in Table 1 all three cell lines can serve as models for human epithelioid MM. As similarities are also high for sarcomatoid MM, also here they may be used as models for. However, AB1, AB22 and AC29 are definitely not models for human biphasic MM.

Methods

Murine MM cell lines

The murine cell lines AB1 and AC29 were obtained from Cell Bank Australia (Westmead, Australia, order #s CBA-0144 and CBA-0152) and AB22 European Collection of Authenticated Cell Cultures (Salisbury, UK—order# ECACC 10092307). For this study, the cells were cultivated and divided into two portions, worked up cytogenetically (portion 1), and used to extract whole-genomic DNA (portion 2) as previously described [[24]].

Molecular cytogenetics

Fluorescence in situ hybridization (FISH) was performed as previously described [[24]]. "SkyPaintTM DNA Kit M-10 for Mouse Chromosomes" (Applied Spectral Imaging, Edingen-Neckarhausen, Germany) was used for multicolor-FISH (mFISH) applying whole chromosome paints, and murine chromosome-specific multicolor banding (mcb) probe mixes for FISH-banding [[31]]. At least 30 metaphases were acquired and analyzed for each probe set on a Zeiss Axioplan microscope, equipped with ISIS software (MetaSystems, Altlussheim, Germany). Array-based comparative genomic hybridization (aCGH) was completed according to standard procedures with "SurePrint G3 Mouse CGH Microarray, 4x180K" (Agilent Technologies, Santa Clara, CA, USA).

Data analysis and translation

The regions of imbalances and breakpoints in AB1, AB22 and AC29 were characterized after analyses of aCGH and mcb data, and aligned with their human homologous regions using Ensembl Genome Browser, as previously described [[24]]. The data we obtained was compared with the literature [[22]] (Tables 1 and 2).

Funding

Open access funding provided by Projekt DEAL. Supported by Grant # 2013.032.1 of the Wilhelm Sander-Stiftung.

Acknowledgements

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

Authors' contributions

TL developed the idea for this study and got funded for it; EW and SA did the practical work and evaluated the FISH-results; MR did the practical work and evaluated the aCGH-results; TL and SA did final evaluation; AS drafted the paper; TL wrote the final version which was approved by all coauthors. All authors read and approved the final manuscript.

Availability of data and materials

All data generated or analysed during this study are included in this published article and its supplementary information files.

Ethics approval and consent to participate

According to the ethical committee (medical faculty) and the Animal Experimentation Commission of the Friedrich Schiller University there are no ethical agreements necessary for studies involving murine tumor cell lines.

Competing interests

The authors declare that they have no competing interests.

Supplementary information

Graph: Additional file 1: Table 1. The regions of gain and loss of copy numbers, as well of breakpoints of balanced rearrangements, observed in AB1, AB22 and AC29 and the corresponding homologue regions in humans, are listed as cytoband and position (GRCh37/hg19).

• aCGH

  • Array comparative genomic hybridization
  • BAP1
  • Breast cancer associated gene 1 (BRCA1) associated protein 1 gene
  • CDKN2A
  • Cyclin-dependent kinase inhibitor 2A gene

• FISH

  • Fluorescence in situ hybridization

• mFISH

  • Multicolor fluorescence in situ hybridization using all 21 murine whole chromosome paints as probes

• MM

  • Malignant mesothelioma

• mcb

  • Murine multicolor banding
  • NF2
  • Neurofibromatosis type 2 gene
  • TP53
  • Tumorsuppressorprotein 53 gene
Supplementary information

Supplementary information accompanies this paper at 10.1186/s13039-020-00511-4.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References 1 Attanoos RL, Pugh MR. The diagnosis of pleural tumors other than mesothelioma. Arch Pathol Lab Med. 2018; 142: 902-913. 10.5858/arpa.2017-0537-RA 2 Opitz I. Management of malignant pleural mesothelioma—the European experience. J Thorac Dis. 2014; 6: S238-S2524032963 3 Dragani TA, Colombo F, Pavlisko EN, Roggli VL. Malignant mesothelioma diagnosed at a younger age is associated with heavier asbestos exposure. Carcinogenesis. 2018; 39: 1151-1156. 10.1093/carcin/bgy089 4 Hiddinga BI, Rolfo C, van Meerbeeck JP. Mesothelioma treatment: Are we on target? A review. J Adv Res. 2015; 6: 319-330. 10.1016/j.jare.2014.11.012 5 Solbes E, Harper RW. Biological responses to asbestos inhalation and pathogenesis of asbestos-related benign and malignant disease. J Investig Med. 2018; 66: 721-727. 10.1136/jim-2017-000628 6 Frost G. The latency period of mesothelioma among a cohort of British asbestos workers (1978–2005). Br J Cancer. 2013; 109: 1965-19733790169. 10.1038/bjc.2013.514 7 Matsumoto S, Nabeshima K, Kamei T, Hiroshima K, Kawahara K, Hata S, Marukawa K, Matsuno Y, Taguchi K, Tsujimura T. Morphology of 9p21 homozygous deletion-positive pleural mesothelioma cells analyzed using fluorescence in situ hybridization and virtual microscope system in effusion cytology. Cancer Cytopathol. 2013; 121: 415-422. 10.1002/cncy.21269 8 Sage AP, Martinez VD, Minatel BC, Pewarchuk ME, Marshall EA, MacAulay GM, Hubaux R, Pearson DD, Goodarzi AA, Dellaire G, Lam WL. Genomics and epigenetics of malignant mesothelioma. High Throughput. 2018; 7: 206163664. 10.3390/ht7030020 9 Chapel DB, Schulte JJ, Husain AN, Krausz T. Application of immunohistochemistry in diagnosis and management of malignant mesothelioma. Transl Lung Cancer Res. 2020; 9: S3-S277082260. 10.21037/tlcr.2019.11.29 Review Sekido Y. Molecular pathogenesis of malignant mesothelioma. Carcinogenesis. 2013; 34: 1413-1419. 10.1093/carcin/bgt166 Yoshikawa Y, Sato A, Tsujimura T, Emi M, Morinaga T, Fukuoka K, Yamada S, Murakami A, Kondo N, Matsumoto S, Okumura Y, Tanaka F, Hasegawa S, Nakano T, Hashimoto-Tamaoki T. Frequent inactivation of the BAP1 gene in epithelioid-type malignant mesothelioma. Cancer Sci. 2012; 103: 868-874. 10.1111/j.1349-7006.2012.02223.x Farzin M, Toon CW, Clarkson A, Sioson L, Watson N, Andrici J, Gill AJ. Loss of expression of BAP1 predicts longer survival in mesothelioma. Pathology. 2015; 47: 302-307. 10.1097/PAT.0000000000000250 Paajanen J, Laaksonen S, Kettunen E, Ilonen I, Vehmas T, Salo J, Räsänen J, Sutinen E, Ollila H, Mäyränpää MI, Myllärniemi M, Wolff H. Histopathological features of epithelioid malignant pleural mesotheliomas in patients with extended survival. Hum Pathol. 2020; 98: 110-119. 10.1016/j.humpath.2020.02.007 De Rienzo A, Testa JR. Malignant mesothelioma. Atlas Genet Cytogenet Oncol Haematol. 2018; 22: 403-406 Hmeljak J, Sanchez-Vega F, Hoadley KA, Shih J, Stewart C, Heiman D, Tarpey P, Danilova L, Drill E, Gibb EA, Bowlby R, Kanchi R, Osmanbeyoglu HU, Sekido Y, Takeshita J, Newton Y, Graim K, Gupta M, Gay CM, Diao L, Gibbs DL, Thorsson V, Iype L, Kantheti H, Severson DT, Ravegnini G, Desmeules P, Jungbluth AA, Travis WD, Dacic S, Chirieac LR, Galateau-Sallé F, Fujimoto J, Husain AN, Silveira HC, Rusch VW, Rintoul RC, Pass H, Kindler H, Zauderer MG, Kwiatkowski DJ, Bueno R, Tsao AS, Creaney J, Lichtenberg T, Leraas K, Bowen JTCGA Research NetworkFelau I, Zenklusen JC, Akbani R, Cherniack AD, Byers LA, Noble MS, Fletcher JA, Robertson AG, Shen R, Aburatani H, Robinson BW, Campbell P, Ladanyi M. Integrative molecular characterization of malignant pleural mesothelioma. Cancer Discov. 2018; 8: 1548-15656310008. 10.1158/2159-8290.CD-18-0804 de Assis LV, Isoldi MC. The function, mechanisms, and role of the genes PTEN and TP53 and the effects of asbestos in the development of malignant mesothelioma: a review focused on the genes' molecular mechanisms. Tumour Biol. 2014; 35: 889-901. 10.1007/s13277-013-1210-4 Patil NS, Righi L, Koeppen H, Zou W, Izzo S, Grosso F, Libener R, Loiacono M, Monica V, Buttigliero C, Novello S, Hegde PS, Papotti M, Kowanetz M, Scagliotti GV. Molecular and histopathological characterization of the tumor immune microenvironment in advanced stage of malignant pleural mesothelioma. J Thorac Oncol. 2018; 13: 124-133. 10.1016/j.jtho.2017.09.1968 Micolucci L, Akhtar MM, Olivieri F, Rippo MR, Procopio AD. Diagnostic value of microRNAs in asbestos exposure and malignant mesothelioma: systematic review and qualitative meta-analysis. Oncotarget. 2016; 7: 58606-586375295457. 10.18632/oncotarget.9686 Boffetta P, Donato F, Pira E, Luu HN, La Vecchia C. Risk of mesothelioma after cessation of asbestos exposure: a systematic review and meta-regression. Int Arch Occup Environ Health. 2019; 92: 949-957. 10.1007/s00420-019-01433-4 Waterston RH, Lindblad-Toh K, Birney E, Rogers J, Abril JF, Agarwal P, Agarwala R, Ainscough R, Alexandersson M, An P, Antonarakis SE, Attwood J, Baertsch R, Bailey J, Barlow K, Beck S, Berry E, Birren B, Bloom T, Bork P, Botcherby M, Bray N, Brent MR, Brown DG, Brown SD, Bult C, Burton J, Butler J, Campbell RD, Carninci P, Cawley S, Chiaromonte F, Chinwalla AT, Church DM, Clamp M, Clee C, Collins FS, Cook LL, Copley RR, Coulson A, Couronne O, Cuff J, Curwen V, Cutts T, Daly M, David R, Davies J, Delehaunty KD, Deri J, Dermitzakis ET, Dewey C, Dickens NJ, Diekhans M, Dodge S, Dubchak I, Dunn DM, Eddy SR, Elnitski L, Emes RD, Eswara P, Eyras E, Felsenfeld A, Fewell GA, Flicek P, Foley K, Frankel WN, Fulton LA, Fulton RS, Furey TS, Gage D, Gibbs RA, Glusman G, Gnerre S, Goldman N, Goodstadt L, Grafham D, Graves TA, Green ED, Gregory S, Guigó R, Guyer M, Hardison RC, Haussler D, Hayashizaki Y, Hillier LW, Hinrichs A, Hlavina W, Holzer T, Hsu F, Hua A, Hubbard T, Hunt A, Jackson I, Jaffe DB, Johnson LS, Jones M, Jones TA, Joy A, Kamal M, Karlsson EK, Karolchik D, Kasprzyk A, Kawai J, Keibler E, Kells C, Kent WJ, Kirby A, Kolbe DL, Korf I, Kucherlapati RS, Kulbokas EJ, Kulp D, Landers T, Leger JP, Leonard S, Letunic I, Levine R, Li J, Li M, Lloyd C, Lucas S, Ma B, Maglott DR, Mardis ER, Matthews L, Mauceli E, Mayer JH, McCarthy M, McCombie WR, McLaren S, McLay K, McPherson JD, Meldrim J, Meredith B, Mesirov JP, Miller W, Miner TL, Mongin E, Montgomery KT, Morgan M, Mott R, Mullikin JC, Muzny DM, Nash WE, Nelson JO, Nhan MN, Nicol R, Ning Z, Nusbaum C, O'Connor MJ, Okazaki Y, Oliver K, Overton-Larty E, Pachter L, Parra G, Pepin KH, Peterson J, Pevzner P, Plumb R, Pohl CS, Poliakov A, Ponce TC, Ponting CP, Potter S, Quail M, Reymond A, Roe BA, Roskin KM, Rubin EM, Rust AG, Santos R, Sapojnikov V, Schultz B, Schultz J, Schwartz MS, Schwartz S, Scott C, Seaman S, Searle S, Sharpe T, Sheridan A, Shownkeen R, Sims S, Singer JB, Slater G, Smit A, Smith DR, Spencer B, Stabenau A, Stange-Thomann N, Sugnet C, Suyama M, Tesler G, Thompson J, Torrents D, Trevaskis E, Tromp J, Ucla C, Ureta-Vidal A, Vinson JP, Von Niederhausern AC, Wade CM, Wall M, Weber RJ, Weiss RB, Wendl MC, West AP, Wetterstrand K, Wheeler R, Whelan S, Wierzbowski J, Willey D, Williams S, Wilson RK, Winter E, Worley KC, Wyman D, Yang S, Yang SP, Zdobnov EM, Zody MC, Lander ES. Initial sequencing and comparative analysis of the mouse genome. Nature. 2002; 420: 520-562. 10.1038/nature01262 Davis MR, Manning LS, Whitaker D, Garlepp MJ, Robinson BWS. Establishment of a murine model of malignant mesothelioma. Int J Cancer. 1992; 52: 881-886. 10.1002/ijc.2910520609 Krismann M, Müller KM, Jaworska M, Johnen G. Molecular cytogenetic differences between histological subtypes of malignant mesotheliomas: DNA cytometry and comparative genomic hybridization of 90 cases. J Pathol. 2002; 197: 363-371. 10.1002/path.1128 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. 2017; 61: 306-312. 10.1369/0022155413476868 Kubicova E, Trifonov V, Borovecki F, Liehr T, Rincic M, Kosyakova N, Hussein SS. 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. 10.2174/1574893611666160606164459 Guja K, Liehr T, Rincic M, Kosyakova N, Hussein Azawi SS. Molecular cytogenetic characterization identified the murine B-Cell lymphoma cell line A-20 as a model for sporadic Burkitt's jymphoma. J Histochem Cytochem. 2017; 65: 669-6775665108. 10.1369/0022155417731319 Rhode H, Liehr T, Kosyakova N, Rincic M, Azawi SSH. Molecular cytogenetic characterization of two murine colorectal cancer cell lines. OBM Genet. 2018; 2: 37. 10.21926/obm.genet.1803037 Steinacker R, Liehr T, Kosyakova N, Rincic M, Hussein Azawi SS. Molecular cytogenetic characterization of two murine cancer cell lines derived from salivary gland. Biol Commun. 2018; 63: 243-255. 10.21638/spbu03.2018.403 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: E4716. 10.3390/ijms21134716 Davoli T, de Lange T. Telomere-driven tetraploidization occurs in human cells undergoing crisis and promotes transformation of mouse cells. Cancer Cell. 2012; 21: 765-7763376354. 10.1016/j.ccr.2012.03.044 Mastromonaco GF, Perrault SD, Betts DH, King WA. Role of chromosome stability and telomere length in the production of viable cell lines for somatic cell nuclear transfer. BMC Dev Biol. 2006; 6: 411590017. 10.1186/1471-213X-6-41 Liehr T, Starke H, Heller A, Kosyakova N, Mrasek K, Gross M, Karst C, Steinhaeuser U, Hunstig F, Fickelscher I, Kuechler A, Trifonov V, Romanenko SA, Weise A. Multicolor fluorescence in situ hybridization (FISH) applied to FISH-banding. Cytogenet Genome Res. 2006; 114: 240-244. 10.1159/000094207

By Eva Wahlbuhl; Thomas Liehr; Martina Rincic and Shaymaa Azawi

Reported by Author; Author; Author; Author

Titel:
Cytogenomic characterization of three murine malignant mesothelioma tumor cell lines
Autor/in / Beteiligte Person: Wahlbuhl, Eva ; Liehr, Thomas ; Rincic, Martina ; Azawi, Shaymaa
Link:
Zeitschrift: Molecular Cytogenetics, Jg. 13 (2020), Heft 1, S. 1-10
Veröffentlichung: BMC, 2020
Medientyp: academicJournal
ISSN: 1755-8166 (print)
DOI: 10.1186/s13039-020-00511-4
Schlagwort:
  • Murine multicolor banding (mcb)
  • Array comparative genomic hybridization (aCGH)
  • Malignant mesothelioma
  • Murine cell line
  • AB1
  • AB22
  • Genetics
  • QH426-470
Sonstiges:
  • Nachgewiesen in: Directory of Open Access Journals
  • Sprachen: English
  • Collection: LCC:Genetics
  • Document Type: article
  • File Description: electronic resource
  • Language: English

Klicken Sie ein Format an und speichern Sie dann die Daten oder geben Sie eine Empfänger-Adresse ein und lassen Sie sich per Email zusenden.

oder
oder

Wählen Sie das für Sie passende Zitationsformat und kopieren Sie es dann in die Zwischenablage, lassen es sich per Mail zusenden oder speichern es als PDF-Datei.

oder
oder

Bitte prüfen Sie, ob die Zitation formal korrekt ist, bevor Sie sie in einer Arbeit verwenden. Benutzen Sie gegebenenfalls den "Exportieren"-Dialog, wenn Sie ein Literaturverwaltungsprogramm verwenden und die Zitat-Angaben selbst formatieren wollen.

xs 0 - 576
sm 576 - 768
md 768 - 992
lg 992 - 1200
xl 1200 - 1366
xxl 1366 -