Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

PVT1 dependence in cancer with MYC copy-number increase

This article has been updated

Abstract

‘Gain’ of supernumerary copies of the 8q24.21 chromosomal region has been shown to be common in many human cancers1,2,3,4,5,6,7,8,9,10,11,12,13 and is associated with poor prognosis7,10,14. The well-characterized myelocytomatosis (MYC) oncogene resides in the 8q24.21 region and is consistently co-gained with an adjacent ‘gene desert’ of approximately 2 megabases that contains the long non-coding RNA gene PVT1, the CCDC26 gene candidate and the GSDMC gene. Whether low copy-number gain of one or more of these genes drives neoplasia is not known. Here we use chromosome engineering in mice to show that a single extra copy of either the Myc gene or the region encompassing Pvt1, Ccdc26 and Gsdmc fails to advance cancer measurably, whereas a single supernumerary segment encompassing all four genes successfully promotes cancer. Gain of PVT1 long non-coding RNA expression was required for high MYC protein levels in 8q24-amplified human cancer cells. PVT1 RNA and MYC protein expression correlated in primary human tumours, and copy number of PVT1 was co-increased in more than 98% of MYC-copy-increase cancers. Ablation of PVT1 from MYC-driven colon cancer line HCT116 diminished its tumorigenic potency. As MYC protein has been refractory to small-molecule inhibition, the dependence of high MYC protein levels on PVT1 long non-coding RNA provides a much needed therapeutic target.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Gain of Myc promotes tumorigenesis only if downstream sequence is co-gained.
Figure 2: Pre-cancerous phenotypes in mouse gain(Myc,Pvt1,Ccdc26,Gsdmc) mammary glands.
Figure 3: Pvt1/PVT1 co-gained with Myc/MYC elevates Myc/MYC protein levels.
Figure 4: PVT1 dependence in MYC-driven tumours.

Similar content being viewed by others

Change history

  • 06 August 2014

    Figure 2a, b, d scale bars were missing and have been added. Minor edits were made to the Fig. 2 legend.

References

  1. Huppi, K., Pitt, J. J., Wahlberg, B. M. & Caplen, N. J. The 8q24 gene desert: an oasis of non-coding transcriptional activity. Front. Genet. 3, 69 (2012)

    Article  Google Scholar 

  2. Haverty, P. M., Hon, L. S., Kaminker, J. S., Chant, J. & Zhang, Z. High-resolution analysis of copy number alterations and associated expression changes in ovarian tumors. BMC Med. Genomics 2, 21 (2009)

    Article  Google Scholar 

  3. Guan, Y. et al. Amplification of PVT1 contributes to the pathophysiology of ovarian and breast cancer. Clin. Cancer Res. 13, 5745–5755 (2007)

    Article  CAS  Google Scholar 

  4. van Duin, M. et al. High-resolution array comparative genomic hybridization of chromosome 8q: evaluation of putative progression markers for gastroesophageal junction adenocarcinomas. Cytogenet. Genome Res. 118, 130–137 (2007)

    Article  CAS  Google Scholar 

  5. Borg, A., Baldetorp, B., Ferno, M., Olsson, H. & Sigurdsson, H. c-myc amplification is an independent prognostic factor in postmenopausal breast cancer. Int. J. Cancer 51, 687–691 (1992)

    Article  CAS  Google Scholar 

  6. Kim, Y. H. et al. Combined microarray analysis of small cell lung cancer reveals altered apoptotic balance and distinct expression signatures of MYC family gene amplification. Oncogene 25, 130–138 (2006)

    Article  CAS  Google Scholar 

  7. Sato, K. et al. Clinical significance of alterations of chromosome 8 in high-grade, advanced, nonmetastatic prostate carcinoma. J. Natl Cancer Inst. 91, 1574–1580 (1999)

    Article  CAS  Google Scholar 

  8. Lapointe, J. et al. Genomic profiling reveals alternative genetic pathways of prostate tumorigenesis. Cancer Res. 67, 8504–8510 (2007)

    Article  CAS  Google Scholar 

  9. Douglas, E. J. et al. Array comparative genomic hybridization analysis of colorectal cancer cell lines and primary carcinomas. Cancer Res. 64, 4817–4825 (2004)

    Article  CAS  Google Scholar 

  10. Zitterbart, K. et al. Low-level copy number changes of MYC genes have a prognostic impact in medulloblastoma. J. Neurooncol. 102, 25–33 (2011)

    Article  CAS  Google Scholar 

  11. Yamada, T. et al. Frequent chromosome 8q gains in human small cell lung carcinoma detected by arbitrarily primed-PCR genomic fingerprinting. Cancer Genet. Cytogenet. 120, 11–17 (2000)

    Article  CAS  Google Scholar 

  12. Le Beau, M. M., Bitts, S., Davis, E. M. & Kogan, S. C. Recurring chromosomal abnormalities in leukemia in PML-RARA transgenic mice parallel human acute promyelocytic leukemia. Blood 99, 2985–2991 (2002)

    Article  CAS  Google Scholar 

  13. Chin, K. et al. Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. Cancer Cell 10, 529–541 (2006)

    Article  MathSciNet  CAS  Google Scholar 

  14. Jain, A. N. et al. Quantitative analysis of chromosomal CGH in human breast tumors associates copy number abnormalities with p53 status and patient survival. Proc. Natl Acad. Sci. USA 98, 7952–7957 (2001)

    Article  ADS  CAS  Google Scholar 

  15. Ramirez-Solis, R., Liu, P. & Bradley, A. Chromosome engineering in mice. Nature 378, 720–724 (1995)

    Article  ADS  CAS  Google Scholar 

  16. Al-Kuraya, K. et al. Prognostic relevance of gene amplifications and coamplifications in breast cancer. Cancer Res. 64, 8534–8540 (2004)

    Article  CAS  Google Scholar 

  17. Park, K., Kwak, K., Kim, J., Lim, S. & Han, S. c-myc amplification is associated with HER2 amplification and closely linked with cell proliferation in tissue microarray of nonselected breast cancers. Hum. Pathol. 36, 634–639 (2005)

    Article  CAS  Google Scholar 

  18. Guy, C. T. et al. Expression of the neu protooncogene in the mammary epithelium of transgenic mice induces metastatic disease. Proc. Natl Acad. Sci. USA 89, 10578–10582 (1992)

    Article  ADS  CAS  Google Scholar 

  19. Saji, S. et al. Estrogen receptors α and β in the rodent mammary gland. Proc. Natl Acad. Sci. USA 97, 337–342 (2000)

    Article  ADS  CAS  Google Scholar 

  20. Carramusa, L. et al. The PVT-1 oncogene is a Myc protein target that is overexpressed in transformed cells. J. Cell. Physiol. 213, 511–518 (2007)

    Article  CAS  Google Scholar 

  21. Lin, M. et al. RNA-Seq of human neurons derived from iPS cells reveals candidate long non-coding RNAs involved in neurogenesis and neuropsychiatric disorders. PLoS ONE 6, e23356 (2011)

    Article  ADS  CAS  Google Scholar 

  22. Zhang, X. et al. Mechanistic insight into Myc stabilization in breast cancer involving aberrant Axin1 expression. Proc. Natl Acad. Sci. USA 109, 2790–2795 (2012)

    Article  ADS  CAS  Google Scholar 

  23. Yeh, E. et al. A signalling pathway controlling c-Myc degradation that impacts oncogenic transformation of human cells. Nature Cell Biol. 6, 308–318 (2004)

    Article  CAS  Google Scholar 

  24. Wang, X. et al. Phosphorylation regulates c-Myc’s oncogenic activity in the mammary gland. Cancer Res. 71, 925–936 (2011)

    Article  CAS  Google Scholar 

  25. Morin, P. J. et al. Activation of β-catenin-Tcf signaling in colon cancer by mutations in β-catenin or APC. Science 275, 1787–1790 (1997)

    Article  CAS  Google Scholar 

  26. Delmore, J. E. et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146, 904–917 (2011)

    Article  CAS  Google Scholar 

  27. Darnell, J. E., Jr Transcription factors as targets for cancer therapy. Nature Rev. Cancer 2, 740–749 (2002)

    Article  CAS  Google Scholar 

  28. Nair, S. K. & Burley, S. K. X-ray structures of Myc-Max and Mad-Max recognizing DNA. Molecular bases of regulation by proto-oncogenic transcription factors. Cell 112, 193–205 (2003)

    Article  CAS  Google Scholar 

  29. Bagchi, A. et al. CHD5 is a tumor suppressor at human 1p36. Cell 128, 459–475 (2007)

    Article  CAS  Google Scholar 

  30. Schwertfeger, K. L. et al. A critical role for the inflammatory response in a mouse model of preneoplastic progression. Cancer Res. 66, 5676–5685 (2006)

    Article  CAS  Google Scholar 

  31. Reed, J. R. et al. Fibroblast growth factor receptor 1 activation in mammary tumor cells promotes macrophage recruitment in a CX3CL1-dependent manner. PLoS ONE 7, e45877 (2012)

    Article  ADS  CAS  Google Scholar 

  32. Lee, G. Y., Kenny, P. A., Lee, E. H. & Bissell, M. J. Three-dimensional culture models of normal and malignant breast epithelial cells. Nature Methods 4, 359–365 (2007)

    Article  CAS  Google Scholar 

  33. Kenny, P. A. et al. The morphologies of breast cancer cell lines in three-dimensional assays correlate with their profiles of gene expression. Mol. Oncol. 1, 84–96 (2007)

    Article  CAS  Google Scholar 

  34. Itou, J. et al. Migration of cardiomyocytes is essential for heart regeneration in zebrafish. Development 139, 4133–4142 (2012)

    Article  CAS  Google Scholar 

  35. Keene, J. D., Komisarow, J. M. & Friedersdorf, M. B. RIP-Chip: the isolation and identification of mRNAs, microRNAs and protein components of ribonucleoprotein complexes from cell extracts. Nature Protocols 1, 302–307 (2006)

    Article  CAS  Google Scholar 

  36. Moriarity, B. S. et al. Simple and efficient methods for enrichment and isolation of endonuclease modified cells. PloS One 9, e96114 (2014)

    Article  ADS  Google Scholar 

  37. Rahrmann, E. P. et al. Forward genetic screen for malignant peripheral nerve sheath tumor formation identifies new genes and pathways driving tumorigenesis. Nature Genet. 45, 756–766 (2013)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank A. T. Vogel for writing statistical analysis scripts; Research Animal Resources, University of Minnesota, for maintaining the mouse colony; S. Horn and L. Oseth for embryonic stem cell blastocyst injection and FISH analysis respectively. This work was supported by Masonic Cancer Center Laboratory start-up funds (to A.B.), and by grants from the Masonic Scholar Award (to A.B.), the Karen Wyckoff Rein in Sarcoma Fund (to A.B.), Translational Workgroup Pilot Project Awards by the Institute of Prostate and Urologic Cancer, University of Minnesota (to A.B.) and an American Cancer Society Institutional Research Grant (award 118198-IRG-58-001-52-IRG92, to A.B.). A.T. was supported by an Indo-US fellowship from the Indo-US Science and Technology Forum.

Author information

Authors and Affiliations

Authors

Contributions

Y.Y.T. and A.B. conceptualized the research programme and designed the experiments; Y.Y.T., B.S.M., H.K., A.T., R.A., P.R., B.R., K.G., T.C.B., J.E., Y.K. and A.B. performed the experiments. Y.Y.T. and W.G. analysed the data; M.G.O. and Y.Y.T. performed the histological analyses; K.L.S., D.A.L., Y.M., Y.K. and A.B. supervised experiments and data analysis; A.B. and Y.M. wrote the manuscript.

Corresponding author

Correspondence to Anindya Bagchi.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Genetic elements shared between human and mouse in the MYCGSDMC interval.

Extended Data Figure 2 Generating the mouse strains.

ac, Chromosome engineering of Hprt-deficient mouse AB2.2 embryonic stem cells was used to develop mouse strains containing an extra copy of Myc (gain(Myc)) (a), Pvt1/Ccdc26/Gsdmc (gain(Pvt1,Ccdc26,Gsdmc)) (b) and Myc/Pvt1/Ccdc26/Gsdmc (gain(Myc,Pvt1,Ccdc26,Gsdmc)) (c). Gene targeting was performed in AB2.2 with targeting vectors obtained from the Mutagenic Insertion and Chromosome Engineering Resource (MICER) that were modified to facilitate detection of correctly targeted clones by PCR. These electroporated embryonic stem cells were cultured in G418 (G, 180 µg ml−1) or puromycine (P, 3 µg ml−1) for 7–10 days. Correctly targeted clones were identified by PCR analysis. Double-targeted embryonic stem cells were electroporated with the transient Cre-recombinase expression vector pOG231. After subsequent selection of recombinants by using hypoxanthine aminopterin thymidime (H) media for 7 days and then recovery of recombinants by using hypoxanthines and thymidine media for 2 days, the HRGRPR clones containing one mouse chromosome 15 with the targeted region duplication (dp) and the other mouse chromosome 15 with the targeted region deletion (df) were identified. d, Summary of gene targeting of mouse genome at the MycL, MycR and GsdmcR loci. e, FISH analysis of gain/loss Myc,Pvt1,Ccdc26,Gsdmc embryonic stem cells with balanced allele for the Myc/Pvt1/Ccdc26/Gsdmc region. Metaphase and interphase preparations from the engineered cells were probed with BAC clone specific for chromosome 15 and located outside (RP23-18H8, red) and within (RP24-78D24, green) the engineered region. The alleles containing the deletion and the duplication are marked.

Extended Data Figure 3 Histopathology of mammary adenocarcinomas and lung metastasis in gain(Myc,Pvt1, Ccdc26,Gsdmc), MMTVneu/+ and gain(Myc,Pvt1,Ccdc26,Gsdmc) respectively.

a, b, Mammary adenocarcinoma in gain(Myc,Pvt1,Ccdc26,Gsdmc),MMTVneu/+ mice. Representative histopathology of mammary tumours from gain(Myc,Pvt1,Ccdc26,Gsdmc),MMTVneu/+ mice. Image shows a solid, expansile tumour that is invading a small blood vessel (arrow; bar, 1000 µm) (a), and numerous mitotic figures (arrows; bar, 100 µm) (b). c, Summary of histopathology showing early onset of mammary adenocarcinoma in gain(Myc,Pvt1,Ccdc26,Gsdmc),MMTVneu/+ mice compared with MMTVneu/+ mice. d, Image of lung metastasis in gain(Myc,Pvt1,Ccdc26,Gsdmc) with spontaneous mammary adenocarcinoma (YYT 528,). e, Histopathology summary of spontaneous, low-penetrance tumour onset in gain(Myc,Pvt1,Ccdc26,Gsdmc) mice.

Extended Data Figure 4 Abnormal oncogenic stress, proliferation and differentiation in gain(Myc,Pvt1,Ccdc26, Gsdmc) mammary ducts.

a, b, Western blot analysis of p53 (a) and phospho-Erk1/2 (b) in total protein lysates from mammary glands of indicated genotypes. The relative densities of p53 and p-ERK1/2 were calculated by normalizing against the GAPDH and total ERK1/2 protein levels respectively. c, Immunofluorescence analysis of ERα (green) on sections of mammary ducts. Cell nuclei positive for ERα are presented as the percentage of total epithelial cell nuclei (DAPI, blue). Images shown are representative three mice per genotype. d, Haematoxylin and eosin staining of the mammary ducts from wild type and gain (M,P,C,G) mice showed precocious alveolar-like phenotype in the latter. This aberrant structure is shown at higher magnification in the right row. e, Immunofluorescence co-staining for DAPI (blue), luminal marker K8 (red) and myoepithelial marker K14 (green) in mice. Arrowheads indicate co-expression of K8 and K14. DAPI-stained nuclei in blue. Mean ± s.e.m. for ac (n = 3). *P < 0.05, ***P < 0.001 by two-tailed Student’s t-test; error bars, s.e.m.

Extended Data Figure 5 Gasdermin expression in mouse mammary tissues.

a, Gsdmc is not expressed in mouse mammary tissue. Semi-quantitative RT–PCR of Gsdmc transcript in mouse colon and mammary tissues. PCR was performed using equal amount of cDNAs derived from colon and mammary tissues, for cycles as indicated. –RT indicates samples treated without reverse transcriptase. β-actin used as a loading control. NC, negative control (water). b–d, Representative RT–qPCR analysis of Gsdmc2 (b), Gsdmc3 (c) and Gsdmc4 (d) mRNA in 10-week-old virgin mouse mammary tissues of all genotypes. Mean ± s.e.m. for bd (n = 3); error bars, s.e.m.

Extended Data Figure 6 PVT1 depletion results into reduction in proliferation in SK-BR-3 and MDA-MB-231 breast cancer cell lines.

a, b, Proliferation assay of human breast cancer cell lines SK-BR-3 (a) and MDA-MB-231 (b) growing in three-dimensional culture after the cell lines were transfected with siCtrl, siMYC, siPVT1 and both (siMYC + siPVT1). Transfection efficiency in each cell line was confirmed as mentioned in the text. Mean ± s.e.m. for a and b (n = 3). **P < 0.01, ***P < 0.001 by two-tailed Student’s t-test. c, d, Inhibition of miRNA expression coded by the PVT1 locus shows no loss of proliferation in SK-BR-3 cells. SK-BR-3 cells transfected with antisense miRNAs were grown in three-dimensional culture as described before. c, Relative expression of individual miRNA in cells treated with its corresponding inhibitor, normalized to the control experiments Each data point represents the mean ± s.e.m. (n = 3, except for miR-1207, where n = 6). d, Percentage of Ki-67 positive cells denotes the proliferation index. Bar graph, mean ± s.e.m. (n = 3); error bars, s.e.m.

Extended Data Figure 7 PVT1 regulates MYC protein level in MDA-MB-231 breast cancer cells.

a, RT–qPCR measurement of MYC (left) and PVT1 (right) RNA levels in MDA-MB-231 cells 48 h after transfection with the indicated siRNAs. b, Reduction in MYC protein in PVT1-depleted MDA-MB-231. Western blot analysis for MYC protein in the total lysates obtained from the MDA-MB-231 cell line transfected with different siRNAs. The relative density for each category was determined by normalizing against the intensity of the GAPDH band. c, Stability assay for MYC protein in MDA-MB-231 cells. Cells were transfected with siCtrl and siPVT1 and then treated with 10 μM cycloheximide for different time points (top panel). The relative density was determined by comparing against the GAPDH level (bottom panel). Mean ± s.e.m. for ac (n = 3). *P < 0.05, **P < 0.01, ***P < 0.001 by two-tailed Student’s t-test. Error bars, s.e.m.

Extended Data Figure 8 PVT1 and MYC co-localize in the SK-BR-3 nuclei.

a, Specificity of fluorescent-labelled anti-PVT1 RNA probe in SK-BR-3 cells. Fluorescent images of SK-BR-3 cells treated with no probe, sense and anti-sense PVT1 RNA probe. DAPI (blue) stain is shown in the lower panel. b, c, Expression and co-localization of MYC and PVT1 in SK-BR-3 nuclei. Representative fluorescent images of SK-BR-3 cells treated with fluorescently labelled anti-MYC antibody (green) and anti-PVT1 RNA (magenta) (b). DAPI indicating cell nuclei is shown in blue. Cells expressing MYC and PVT1 are indicated by red arrows whereas those expressing low levels of MYC and PVT1 are indicated by white arrows. Merge panel represents overlapping images of DAPI, MYC and PVT1 panels. The nuclei with co-localization of MYC and PVT1 are shown by red arrows. c, Quantification of SK-BR-3 nuclei with co-localization of MYC and PVT1 (n = 87).

Extended Data Figure 9 Incidence MYC and PVT1 co-gain in human cancers.

a, Number of 8q24 gain-associated human cancers showing gain of MYC but not PVT1 (blue), gain of PVT1 but not MYC (orange) and co-gain of MYC + PVT1 (green) in TCGA (left) and Progenetix copy-number database (right). b, Pie chart showing breast cancer samples in TCGA expressing high HER2 transcript with or without co-gain of MYC + PVT1. c, The ratio of MYC + PVT1 + CCDC26 and MYC + PVT1 + GSDMC co-amplification among MYC + PVT1 co-gained TCGA samples at different amplification levels (segment mean cutoff). d, The ratio of MYC + PVT1 co-amplification among MYC-gained cancers on different segment mean cutoffs. e, f, Tissue microarray analysis of PVT1 RNA and MYC protein expression in primary human tumours. Images of 32 primary human tumours showing in situ hybridization using digoxigenin-labelled PVT1 probe (purple, top panel) and MYC immunohistochemistry using anti-MYC antibody (brown, bottom panel) (e). f, Specification of multiple organ normal and diseased tissue microarray, single core per case, eight types of tumour (breast, colon, oesophagus, kidney, liver, lung, rectum, stomach) (BC00119).

Extended Data Figure 10 Generating the ΔPVT1 HCT116.

a, Schematic representation of CRISPR-mediated deletion of 307 kilobase PVT1 gene in HCT116. Black triangles denote CRISPR specific for upstream of exon 1 and downstream of exon 8 of PVT1. PCR primers F1, F2 and R1 are denoted. b, A CRISPR-mediated deletion of PVT1PVT1) can be detected by PCR amplicon using primers F1 and R1, whereas the control with PVT1 locus intact (PVT1+) can be screened by using F2 and R1. c, RT–qPCR analysis of PVT1 transcript in PVT1+ and ΔPVT1 HCT116 cells (n = 3). d, Relative cellular proliferation abilities of PVT1+ and ΔPVT1 HCT116 cells were evaluated by MTS assays. Data are mean ± s.e.m. for c and d (n = 3). *P < 0.05, **P < 0.01, ***P < 0.001 by two-tailed Student’s t-test. Error bars, s.e.m. e, Luciferase-based images of the presence of tumour lesions detected 3–17 days after subcutaneous implantation of PVT1+ and ΔPVT1 HCT116 cells in nude mice.

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tseng, YY., Moriarity, B., Gong, W. et al. PVT1 dependence in cancer with MYC copy-number increase. Nature 512, 82–86 (2014). https://doi.org/10.1038/nature13311

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13311

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer