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.

  • Article
  • Published:

RASSF1A–LATS1 signalling stabilizes replication forks by restricting CDK2-mediated phosphorylation of BRCA2

An Erratum to this article was published on 27 March 2015

This article has been updated

Abstract

Genomic instability is a key hallmark of cancer leading to tumour heterogeneity and therapeutic resistance. BRCA2 has a fundamental role in error-free DNA repair but also sustains genome integrity by promoting RAD51 nucleofilament formation at stalled replication forks. CDK2 phosphorylates BRCA2 (pS3291-BRCA2) to limit stabilizing contacts with polymerized RAD51; however, how replication stress modulates CDK2 activity and whether loss of pS3291-BRCA2 regulation results in genomic instability of tumours are not known. Here we demonstrate that the Hippo pathway kinase LATS1 interacts with CDK2 in response to genotoxic stress to constrain pS3291-BRCA2 and support RAD51 nucleofilaments, thereby maintaining genomic fidelity during replication stalling. We also show that LATS1 forms part of an ATR-mediated response to replication stress that requires the tumour suppressor RASSF1A. Importantly, perturbation of the ATR–RASSF1A–LATS1 signalling axis leads to genomic defects associated with loss of BRCA2 function and contributes to genomic instability and ‘BRCA-ness’ in lung cancers.

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: LATS1 regulates RAD51 nucleofilament formation in response to replication stress in an homologous-recombination-independent manner.
Figure 2: LATS1 interacts with CDK2 in response to genotoxic stress modulating its kinase activity towards BRCA2.
Figure 3: Tumour suppressor RASSF1A stimulates LATS1–CDK2 interaction in response to ATR activation.
Figure 4: Deletion of the RASSF1A–LATS1 axis compromises the stability of nascent DNA at stalled forks.
Figure 5: Deletion of the RASSF1A–LATS1 axis induces chromosomal aberrations.
Figure 6: RASSF1A methylation correlates with increased CNV in lung cancer patients.
Figure 7: Model of RASSF1A–LATS1–CDK2 signalling and the protection of stalled replication forks.

Similar content being viewed by others

Change history

  • 11 March 2015

    In the original version of this Article, the bar chart in Fig. 6 was incorrect. This has now been corrected in all online versions of this Article.

References

  1. Olcina, M. M. et al. Replication stress and chromatin context link ATM activation to a role in DNA replication. Mol. Cell 52, 758–766 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Petermann, E. & Helleday, T. Pathways of mammalian replication fork restart. Nat. Rev. Mol. Cell Biol. 11, 683–687 (2010).

    Article  CAS  PubMed  Google Scholar 

  3. Negrini, S., Gorgoulis, V. G. & Halazonetis, T. D. Genomic instability—an evolving hallmark of cancer. Nat. Rev. Mol. Cell Biol. 11, 220–228 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Murga, M. et al. Exploiting oncogene-induced replicative stress for the selective killing of Myc-driven tumors. Nat. Struct. Mol. Biol. 18, 1331–1335 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bartek, J. & Lukas, J. DNA damage checkpoints: from initiation to recovery or adaptation. Curr. Opin. Cell Biol. 19, 238–245 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Halazonetis, T. D., Gorgoulis, V. G. & Bartek, J. An oncogene-induced DNA damage model for cancer development. Science 319, 1352–1355 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Helmrich, A., Ballarino, M., Nudler, E. & Tora, L. Transcription-replication encounters, consequences and genomic instability. Nat. Struct. Mol. Biol. 20, 412–418 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Schlacher, K. et al. Double-strand break repair-independent role for BRCA2 in blocking stalled replication fork degradation by MRE11. Cell 145, 529–542 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Schlacher, K., Wu, H. & Jasin, M. A distinct replication fork protection pathway connects Fanconi anemia tumor suppressors to RAD51-BRCA1/2. Cancer Cell 22, 106–116 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hashimoto, Y., Ray Chaudhuri, A., Lopes, M. & Costanzo, V. Rad51 protects nascent DNA from Mre11-dependent degradation and promotes continuous DNA synthesis. Nat. Struct. Mol. Biol. 17, 1305–1311 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Ying, S., Hamdy, F. C. & Helleday, T. Mre11-dependent degradation of stalled DNA replication forks is prevented by BRCA2 and PARP1. Cancer Res. 72, 2814–2821 (2012).

    Article  CAS  PubMed  Google Scholar 

  12. Petermann, E., Orta, M. L., Issaeva, N., Schultz, N. & Helleday, T. Hydroxyurea-stalled replication forks become progressively inactivated and require two different RAD51-mediated pathways for restart and repair. Mol. Cell 37, 492–502 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hashimoto, Y., Puddu, F. & Costanzo, V. RAD51- and MRE11-dependent reassembly of uncoupled CMG helicase complex at collapsed replication forks. Nat. Struct. Mol. Biol. 19, 17–24 (2012).

    Article  CAS  Google Scholar 

  14. Esashi, F., Galkin, V. E., Yu, X., Egelman, E. H. & West, S. C. Stabilization of RAD51 nucleoprotein filaments by the C-terminal region of BRCA2. Nat. Struct. Mol. Biol. 14, 468–474 (2007).

    Article  CAS  PubMed  Google Scholar 

  15. Davies, O. R. & Pellegrini, L. Interaction with the BRCA2 C terminus protects RAD51-DNA filaments from disassembly by BRC repeats. Nat. Struct. Mol. Biol. 14, 475–483 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Esashi, F. et al. CDK-dependent phosphorylation of BRCA2 as a regulatory mechanism for recombinational repair. Nature 434, 598–604 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Pan, D. The hippo signaling pathway in development and cancer. Dev. Cell 19, 491–505 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Zeng, Q. & Hong, W. The emerging role of the hippo pathway in cell contact inhibition, organ size control, and cancer development in mammals. Cancer Cell 13, 188–192 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. Visser, S. & Yang, X. LATS tumor suppressor: a new governor of cellular homeostasis. Cell Cycle 9, 3892–3903 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Hamilton, G., Yee, K. S., Scrace, S. & O’Neill, E. ATM regulates a RASSF1A-dependent DNA damage response. Curr. Biol. 19, 2020–2025 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Yee, K. S. et al. A RASSF1A polymorphism restricts p53/p73 activation and associates with poor survival and accelerated age of onset of soft tissue sarcoma. Cancer Res. 72, 2206–2217 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kanzaki, H. et al. Single nucleotide polymorphism at codon 133 of the RASSF1 gene is preferentially associated with human lung adenocarcinoma risk. Cancer Lett. 238, 128–134 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. Gao, B. et al. RASSF1A polymorphism A133S is associated with early onset breast cancer in BRCA1/2 mutation carriers. Cancer Res. 68, 22–25 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bergqvist, J. et al. RASSF1A polymorphism in familial breast cancer. Fam. Cancer 9, 263–265 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Wang, J., Wang, B., Chen, X. & Bi, J. The prognostic value of RASSF1A promoter hypermethylation in non-small cell lung carcinoma: a systematic review and meta-analysis. Carcinogenesis 32, 411–416 (2011).

    Article  CAS  PubMed  Google Scholar 

  26. Jiang, Y., Cui, L., Chen, W. D., Shen, S. H. & Ding, L. D. The prognostic role of RASSF1A promoter methylation in breast cancer: a meta-analysis of published data. PLoS ONE 7, e36780 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Burbee, D. G. et al. Epigenetic inactivation of RASSF1A in lung and breast cancers and malignant phenotype suppression. J. Natl Cancer Inst. 93, 691–699 (2001).

    Article  CAS  PubMed  Google Scholar 

  28. Matsuoka, S. et al. ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science 316, 1160–1166 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Cimprich, K. A. & Cortez, D. ATR: an essential regulator of genome integrity. Nat. Rev. Mol. Cell Biol. 9, 616–627 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Vispe, S., Cazaux, C., Lesca, C. & Defais, M. Overexpression of Rad51 protein stimulates homologous recombination and increases resistance of mammalian cells to ionizing radiation. Nucleic Acids Res. 26, 2859–2864 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Siaud, N. et al. Plasticity of BRCA2 function in homologous recombination: genetic interactions of the PALB2 and DNA binding domains. PLoS Genet. 7, e1002409 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Feng, Z. & Zhang, J. A dual role of BRCA1 in two distinct homologous recombination mediated repair in response to replication arrest. Nucleic Acids Res. 40, 726–738 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. Hanada, K. et al. The structure-specific endonuclease Mus81 contributes to replication restart by generating double-strand DNA breaks. Nat. Struct. Mol. Biol. 14, 1096–1104 (2007).

    Article  CAS  PubMed  Google Scholar 

  34. Dong, J. et al. Elucidation of a universal size-control mechanism in Drosophila and mammals. Cell 130, 1120–1133 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. St John, M. A. et al. Mice deficient of Lats1 develop soft-tissue sarcomas, ovarian tumours and pituitary dysfunction. Nat. Genet. 21, 182–186 (1999).

    Article  CAS  PubMed  Google Scholar 

  36. Camargo, F. D. et al. YAP1 increases organ size and expands undifferentiated progenitor cells. Curr. Biol. 17, 2054–2060 (2007).

    Article  CAS  PubMed  Google Scholar 

  37. Tao, W. et al. Human homologue of the Drosophila melanogaster lats tumour suppressor modulates CDC2 activity. Nat. Genet. 21, 177–181 (1999).

    Article  CAS  PubMed  Google Scholar 

  38. Li, Y. et al. Lats2, a putative tumor suppressor, inhibits G1/S transition. Oncogene 22, 4398–4405 (2003).

    Article  CAS  PubMed  Google Scholar 

  39. Yabuta, N. et al. N-terminal truncation of Lats1 causes abnormal cell growth control and chromosomal instability. J. Cell Sci. 126, 508–520 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Ayoub, N. et al. The carboxyl terminus of Brca2 links the disassembly of Rad51 complexes to mitotic entry. Current Biol. 19, 1075–1085 (2009).

    Article  CAS  Google Scholar 

  41. Matallanas, D. et al. RASSF1A elicits apoptosis through an MST2 pathway directing proapoptotic transcription by the p73 tumor suppressor protein. Mol. Cell 27, 962–975 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Traven, A. & Heierhorst, J. SQ/TQ cluster domains: concentrated ATM/ATR kinase phosphorylation site regions in DNA-damage-response proteins. BioEssays 27, 397–407 (2005).

    Article  CAS  PubMed  Google Scholar 

  43. Lopez-Contreras, A. J. & Fernandez-Capetillo, O. The ATR barrier to replication-born DNA damage. DNA Repair 9, 1249–1255 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chaudhury, I., Sareen, A., Raghunandan, M. & Sobeck, A. FANCD2 regulates BLM complex functions independently of FANCI to promote replication fork recovery. Nucleic Acids Res. 41, 6444–6459 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Song, M. S. et al. The tumour suppressor RASSF1A regulates mitosis by inhibiting the APC–Cdc20 complex. Nat. Cell Biol. 6, 129–137 (2004).

    Article  CAS  PubMed  Google Scholar 

  46. Endoh, H. et al. RASSF1A gene inactivation in non-small cell lung cancer and its clinical implication. Int. J. Cancer 106, 45–51 (2003).

    Article  CAS  PubMed  Google Scholar 

  47. Matallanas, D. et al. Mutant K-Ras activation of the proapoptotic MST2 pathway is antagonized by wild-type K-Ras. Mol. Cell 44, 893–906 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. O’Neill, E., Rushworth, L., Baccarini, M. & Kolch, W. Role of the kinase MST2 in suppression of apoptosis by the proto-oncogene product Raf-1. Science 306, 2267–2270 (2004).

    Article  PubMed  Google Scholar 

  49. Romano, D. et al. Protein interaction switches coordinate Raf-1 and MST2/Hippo signalling. Nat. Cell Biol. 16, 673–684 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Van der Weyden, L. et al. Loss of RASSF1A synergizes with deregulated RUNX2 signaling in tumorigenesis. Cancer Res. 72, 3817–3827 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kapoor, A. et al. Yap1 activation enables bypass of oncogenic kras addiction in pancreatic cancer. Cell 158, 185–197 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Shao, D. D. et al. KRAS and YAP1 converge to regulate EMT and tumor survival. Cell 158, 171–184 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wang, J. et al. Value of p16INK4a and RASSF1A promoter hypermethylation in prognosis of patients with resectable non-small cell lung cancer. Clin. Cancer Res. 10, 6119–6125 (2004).

    Article  CAS  PubMed  Google Scholar 

  54. Tanemura, A. et al. CpG island methylator phenotype predicts progression of malignant melanoma. Clin. Cancer Res. 15, 1801–1807 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. De Fraipont, F. et al. An apoptosis methylation prognostic signature for early lung cancer in the IFCT-0002 trial. Clin. Cancer Res. 18, 2976–2986 (2012).

    Article  CAS  PubMed  Google Scholar 

  56. Van der Weyden, L. et al. The RASSF1A isoform of RASSF1 promotes microtubule stability and suppresses tumorigenesis. Mol. Cell. Biol. 25, 8356–8367 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Chan, E. H. et al. The Ste20-like kinase Mst2 activates the human large tumor suppressor kinase Lats1. Oncogene 24, 2076–2086 (2005).

    Article  CAS  PubMed  Google Scholar 

  58. Khoronenkova, S. V. et al. ATM-dependent downregulation of USP7/HAUSP by PPM1G activates p53 response to DNA damage. Mol. Cell 45, 801–813 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  PubMed  Google Scholar 

  60. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge T. Helleday for helpful discussions, M. Woodcock for fluorescence-activated cell sorting analysis and A. G. Abraham for useful comments on the manuscript. Lats1−/− MEFs were provided by T. Xu and myc–LATS1 constructs by H. Silljé. This work was funded by Cancer Research UK A19277, Vertex and the Medical Research Council (MRC).

Author information

Authors and Affiliations

Authors

Contributions

D-E.P. designed and performed most of the experiments, analysed data and contributed to writing the paper; R.L. performed initial experiments for LATS1–CDK2 interaction, γH2AX analysis, pBRCA2 western blots and I-SceI assay; I.P. transferred the DNA fibre technology; K.S.Y. performed initial immunoprecipitation experiments for RASSF1A effect on LATS1–CDK2 interaction. G.H. performed the in vitro kinase assays; A.M.G. performed the TCGA analysis; L.v.d.W. isolated Rassf1A−/− mouse embryonic fibroblasts; F.E. provided BRCA2 reagents and advice; E.M.H. helped with reagents and advice; E.O’N. conceived, coordinated and supervised the project, designed experiments, analysed data and wrote the paper.

Corresponding author

Correspondence to Eric O’Neill.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 LATS1 ablation results in defective repair kinetics and induces a G2/M arrest.

(a) DNA content analysis at the indicated time points of U2OS cells treated with siRNA against LATS1 or control siRNA after exposure to 4 Gy Ionizing Radiation (γIR). The percentage of cells at the different phases of the cell cycle was quantified and presented. (b) Quantification of γH2AX foci in Lats1+/+, Lats1−/−, Lats1−/−mycLATS1, Lats1−/−mycLats1KD MEFs after exposure to 10 Gy γIR at the indicated time points. Bar-graphs indicate the percentage (%) of cells stained positive for γH2AX in 15 fields of view from a representative experiment out of n = 3 experiments. Error bars indicate Standard Error of the Mean (s.e.m.). Statistical significance was determined by a two-tailed t-test. P < 0.05 (cLats1+/+, Lats1−/− and Lats1−/− cells expressing wt hLATS1 (Lats1−/−mycLATS1) or a hLATS1 kinase dead derivative LATS1-D846A (Lats1−/−LATS1KD) were treated with 10 Gy γIR. Total cell extracts were isolated at the indicated time points after irradiation and probed with the indicated antibodies (full panel of antibodies for Fig. 1b) (d,e) U2OS cells were transfected with siRNA against LATS1 or Non Targeting siRNAi (siNT) and exposed to 4 Gy γIR. Cells were either harvested at the indicated time points after irradiation and analysed with Western Blotting for the expression of the indicated antibodies (d) or fixed and processed for immunofluorescence for γH2AX foci formation (e). Bargrahs derived from 15 fields of view from a representative experiment and error bars indicate s.e.m. Statistical significance was determined by a two-tailed t-test, P < 0.05. (f) Lats1+/+, Lats1−/−, Lats1−/−mycLATS1 and Lats1−/−hmycLATS1KD MEFs were treated with 10 Gy γIR and after the indicated time points single-stranded DNA fragments were separated from matured DNA by alkaline single cell electrophoresis. Quantification of the percentage of comet tail DNA after alkaline comet assay for each condition is shown next to representative images. 50 cells were counted per condition in n = 3 independent experiments. Error bars derive from n = 3 independent experiments and represent standard deviation. Statistical significance was determined by a two-tailed t-test. P < 0.05.

Supplementary Figure 2 LATS1 is necessary for the establishment of RAD51 foci in response to genotoxic stress, independently of YAP.

(a) Representative plots of two-colour fluorescence analysis for U2OS cells stably transfected with DR-GFP (related to Fig. 1c). The percentage of green fluorescent cells falling above the diagonal for each transfection is indicated. (FL1) Green fluorescence; (FL2) orange fluorescence. (bLats1+/+, Lats1−/−, Lats1−/−mycLATS1 and Lats1−/−mycLATS1KD MEFs were exposed to 10 Gy γIR. After the indicated time points cells were fixed and assessed for RAD51 foci by immunofluorescence. The percentage of RAD51 positive cells (> than 9 RAD51 foci/cell) is presented. (c) Quantification graphs and representative pictures of RAD51 foci in Lats1−/− MEFs or WT MEFs treated with siNT, siMST2 or siYAP after 4 h exposure to 2 mM HU are shown. Error bars represent standard deviation from n = 3 independent experiments. Statistical significance was determined by a two-tailed t-test P < 0.05,P < 0.01. Scale bars, 10 μm.

Supplementary Figure 3 LATS1 compromises CDK2 kinase activity to S3291-BRCA2 in response to stress.

(a) Detection of pS3291-BRCA2 in lysates of HT1080 cells treated with the indicated siRNAs and expressing Myc-LATS1 or control plasmid. Line graphs represent time course measurement of pS3291-BRCA2 levels at indicated times post 4 Gy γIR in cycling cells (bottom) or nocodozol treated cells (top). In pS3291-BRCA2 quantification plots, error bars represent the variation in the densitometry of the representative image. Quantification of cycling cells also presented in Fig. 2e in a different scale. (b) In vitro kinase assay of immunoprecipitated CDK2 from Lats1+/+ and Lats1−/− MEFs incubated with C-terminal BRCA2 purified peptide (His-TR2) in the presence of 32[P]ATP. Reactions were resolved by SDS polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to polyvinylidene fluoride (PVDF) membrane. The membrane was visualized by phosphoimage detection (PMI imager, Bio-Rad) and probed with the indicated antibodies.

Supplementary Figure 4 RASSF1A/LATS1 signalling is necessary for the stability of nascent DNA during replication stress.

(a,b) CIdU tract length distributions and representative pictures from DNA fibres from Lats1+/+ and Lats1−/− MEFs (a) and Rassf1A+/+, Rassf1A−/− MEFs (b). Sketch above delineates experimental design. Bar-graphs derived from a representative experiment. 100 tracks were analysed per condition. Mean track lengths and standard deviation given in parenthesis derived from n = 3 independent experiments. Statistical significance was determined by a two-tailed t-test. (c) Examples of DNA fibre images from the indicated cells and conditions. The length of CIdU and IdU tracts are presented in Fig. 4a and Supplementary Fig. 5. (d) Representative images from Rassf1A+/+, Rassf1A−/−, Rassf1A−/−FLAGR1A and Rassf1A−/−FLAG−R1A−A133S MEFs. Length of CIdU and IdU tracts were analysed in Fig. 4c and Supplementary Fig. 5. Scale bar, 10 μm.

Supplementary Figure 5 Deletion of RASSF1A/LATS1 axis compromises fork recovery after stress.

(a) IdU tract length distributions from DNA fibres of Lats1+/+, Lats1−/− and Lats1−/−mycLATS1 after release from 4 h treatment with 2 mM HU, to determine the ability of the cells to recover after replication stress. (b) IdU tract length distributions of Rassf1A+/+, Rassf1A−/−, Rassf1A−/−FLAG−R1A and Rassf1A−/−FLAG−R1A−A133S MEFs after release from 2 mM HU. 100 tracks were analysed in each condition in n = 3 independent experiments. Statistical significance was determined by a two-tailed t-test. Bar-graphs derived from a representative experiment. 100 tracks were analysed per condition. Mean track lengths and standard deviation given in parenthesis derived from n = 3 independent experiments. Statistical significance was determined by a two-tailed t-test. Sketch delineates experimental design.

Supplementary Figure 6 Chromosomal instability accumulates after exposure to replication stress in the absence of RASSF1A/LATS1 signalling.

(a) Representative pictures from metaphase spreads from Lats1+/+, Lats1−/−, Rassf1A+/+ and Rassf1A−/− untreated MEFs quantified for chromosomal abnormalities on Fig. 5a, b. (b) Western Blot analysis for LATS1 and RASSF1A levels in U2OS cells that were examined for chromosomal aberrations in Fig. 5c. (c) Western Blot analysis to assess LATS1 depletion and FLAG-R1A overexpression in H1299 cells used for metaphase spreads in Fig. 5d. (d) U2OS cells treated with Non Targeting siRNA, siRNA against LATS1 or siRNA against RASSF1A were treated or not with HU and released for 16 h prior fixation. DNA was stained with DAPI and cells positive for micronuclei (MN) were scored. 300 cells were scored per condition in n = 3 independent experiments. Error bars represent standard deviation. Statistical significance was determined by a two-tailed t-test. Scale bar, 10 μm.P < 0.01,P < 0.001.

Supplementary Figure 7 Loss of RASSF1A does not correlate with increased mutation frequency.

Correlation of RASSF1 promoter methylation (assessed with illumina HM450) with genomic mutation count in lung adenocarcinoma datasets from the cancer genome atlas database (TCGA, Provisional). A total of 115 patients with Lung Adenocarcinoma were analysed using the Fisher’s exact test. Absolute numbers and P values are presented in the table.

Supplementary Figure 8 Original uncropped images of western blots.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1344 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pefani, DE., Latusek, R., Pires, I. et al. RASSF1A–LATS1 signalling stabilizes replication forks by restricting CDK2-mediated phosphorylation of BRCA2. Nat Cell Biol 16, 962–971 (2014). https://doi.org/10.1038/ncb3035

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3035

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing