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:

Low lamin A levels enhance confined cell migration and metastatic capacity in breast cancer

Abstract

Aberrations in nuclear size and shape are commonly used to identify cancerous tissue. However, it remains unclear whether the disturbed nuclear structure directly contributes to the cancer pathology or is merely a consequence of other events occurring during tumorigenesis. Here, we show that highly invasive and proliferative breast cancer cells frequently exhibit Akt-driven lower expression of the nuclear envelope proteins lamin A/C, leading to increased nuclear deformability that permits enhanced cell migration through confined environments that mimic interstitial spaces encountered during metastasis. Importantly, increasing lamin A/C expression in highly invasive breast cancer cells reflected gene expression changes characteristic of human breast tumors with higher LMNA expression, and specifically affected pathways related to cell-ECM interactions, cell metabolism, and PI3K/Akt signaling. Further supporting an important role of lamins in breast cancer metastasis, analysis of lamin levels in human breast tumors revealed a significant association between lower lamin A levels, Akt signaling, and decreased disease-free survival. These findings suggest that downregulation of lamin A/C in breast cancer cells may influence both cellular physical properties and biochemical signaling to promote metastatic progression.

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

Fig. 1: Nuclear size and stiffness vary widely across breast cancer cell lines and correspond to lamin A/C levels.
Fig. 2: Decreased A-type lamin levels result in the enhanced nuclear deformability of invasive breast cancer cells.
Fig. 3: Decreased Lamin A levels facilitate migration through confined spaces.
Fig. 4: Breast cancer cells with increased metastatic capacity have reduced lamin A expression.
Fig. 5: Increasing lamin A in breast cancer cells alters expression of proteins involved in cell metabolism, extracellular remodeling, adhesion, and cytoskeleton dynamics.
Fig. 6: Lamin A levels modulate breast cancer cell morphology and proliferation.
Fig. 7: Decreased A-type lamin levels are associated with increased proliferation and Akt signaling in human breast tumors.
Fig. 8: Decreased lamin A levels are associated with worse prognosis in breast cancer.

Similar content being viewed by others

References

  1. de Las Heras JI, Schirmer EC. The nuclear envelope and cancer: a diagnostic perspective and historical overview. Adv Exp Med Biol. 2014;773:5–26.

    Article  PubMed  Google Scholar 

  2. Caille N, Thoumine O, Tardy Y, Meister JJ. Contribution of the nucleus to the mechanical properties of endothelial cells. J Biomech. 2002;35:177–87.

    Article  PubMed  Google Scholar 

  3. Guilak F, Tedrow JR, Burgkart R. Viscoelastic properties of the cell nucleus. Biochem Biophys Res Commun. 2000;269:781–6.

    Article  CAS  PubMed  Google Scholar 

  4. Denais C, Lammerding J. Nuclear mechanics in cancer. Adv Exp Med Biol. 2014;773:435–70.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Tseng Y, Lee JS, Kole TP, Jiang I, Wirtz D. Micro-organization and visco-elasticity of the interphase nucleus revealed by particle nanotracking. J Cell Sci. 2004;117:2159–67.

    Article  CAS  PubMed  Google Scholar 

  6. Wolf K, Te Lindert M, Krause M, Alexander S, Te Riet J, Willis AL, et al. Physical limits of cell migration: control by ECM space and nuclear deformation and tuning by proteolysis and traction force. J Cell Biol. 2013;201:1069–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Stoitzner P, Pfaller K, Stossel H, Romani N. A close-up view of migrating Langerhans cells in the skin. J Investig Dermatol. 2002;118:117–25.

    Article  CAS  PubMed  Google Scholar 

  8. Weigelin B, Bakker GJ, Friedl P. Intravital third harmonic generation microscopy of collective melanoma cell invasion. IntraVital. 2012;1:32–43.

    Article  PubMed  Google Scholar 

  9. Doerschuk CM, Beyers N, Coxson HO, Wiggs B, Hogg JC. Comparison of neutrophil and capillary diameters and their relation to neutrophil sequestration in the lung. J Appl Physiol. 1993;74:3040–5.

    Article  CAS  PubMed  Google Scholar 

  10. Davidson PM, Denais C, Bakshi MC, Lammerding J. Nuclear deformability constitutes a rate-limiting step during cell migration in 3-D environments. Cell Mol Bioeng. 2014;7:293–306.

    Article  CAS  PubMed  Google Scholar 

  11. Harada T, Swift J, Irianto J, Shin JW, Spinler KR, Athirasala A, et al. Nuclear lamin stiffness is a barrier to 3D migration, but softness can limit survival. J Cell Biol. 2014;204:669–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lautscham LA, Kammerer C, Lange JR, Kolb T, Mark C, Schilling A, et al. Migration in confined 3D environments is determined by a combination of adhesiveness, nuclear volume, contractility, and cell stiffness. Biophys J. 2015;109:900–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Mukherjee A, Barai A, Singh RK, Yan W, Sen S. Nuclear plasticity increases susceptibility to damage during confined migration. PLoS Comput Biol. 2020;16:e1008300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bustin M, Misteli T. Nongenetic functions of the genome. Science. 2016;352:aad6933.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Reddy KL, Feinberg AP. Higher order chromatin organization in cancer. Semin Cancer Biol. 2013;23:109–15.

    Article  CAS  PubMed  Google Scholar 

  16. Bell ES, Lammerding J. Causes and consequences of nuclear envelope alterations in tumour progression. Eur J Cell Biol. 2016;95:449–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Irianto J, Pfeifer CR, Ivanovska IL, Swift J, Discher D. Nuclear lamins in cancer. Cell Mol Bioeng. 2016;9:258–67.

    Article  CAS  PubMed  Google Scholar 

  18. Pombo A, Dillon N. Three-dimensional genome architecture: players and mechanisms. Nat Rev Mol Cell Biol. 2015;16:245–57.

    Article  CAS  PubMed  Google Scholar 

  19. Hetzer MW. The nuclear envelope. Cold Spring Harb Perspect Biol. 2010;2:a000539.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Stephens AD, Banigan EJ, Adam SA, Goldman RD, Marko JF. Chromatin and lamin A determine two different mechanical response regimes of the cell nucleus. Mol Biol Cell. 2017;28:1984–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Stephens AD, Liu PZ, Banigan EJ, Almassalha LM, Backman V, Adam SA, et al. Chromatin histone modifications and rigidity affect nuclear morphology independent of lamins. Mol Biol Cell. 2018;29:220–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Schape J, Prausse S, Radmacher M, Stick R. Influence of lamin A on the mechanical properties of amphibian oocyte nuclei measured by atomic force microscopy. Biophys J. 2009;96:4319–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Swift J, Ivanovska IL, Buxboim A, Harada T, Dingal PC, Pinter J, et al. Nuclear lamin-A scales with tissue stiffness and enhances matrix-directed differentiation. Science. 2013;341:1240104.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Lammerding J, Fong LG, Ji JY, Reue K, Stewart CL, Young SG, et al. Lamins A and C but not lamin B1 regulate nuclear mechanics. J Biol Chem. 2006;281:25768–80.

    Article  CAS  PubMed  Google Scholar 

  25. Lammerding J, Schulze PC, Takahashi T, Kozlov S, Sullivan T, Kamm RD, et al. Lamin A/C deficiency causes defective nuclear mechanics and mechanotransduction. J Clin Investig. 2004;113:370–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zwerger M, Jaalouk DE, Lombardi ML, Isermann P, Mauermann M, Dialynas G, et al. Myopathic lamin mutations impair nuclear stability in cells and tissue and disrupt nucleo-cytoskeletal coupling. Hum Mol Genet. 2013;22:2335–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Earle AJ, Kirby TJ, Fedorchak GR, Isermann P, Patel J, Iruvanti S, et al. Mutant lamins cause nuclear envelope rupture and DNA damage in skeletal muscle cells. Nat Mater. 2020;19:464–73.

    Article  CAS  PubMed  Google Scholar 

  28. Cho S, Vashisth M, Abbas A, Majkut S, Vogel K, Xia Y, et al. Mechanosensing by the lamina protects against nuclear rupture, DNA damage, and cell-cycle arrest. Dev Cell. 2019;49:920–35.e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Vargas JD, Hatch EM, Anderson DJ, Hetzer MW. Transient nuclear envelope rupturing during interphase in human cancer cells. Nucleus. 2012;3:88–100.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Denais CM, Gilbert RM, Isermann P, McGregor AL, Te Lindert M, Weigelin B, et al. Nuclear envelope rupture and repair during cancer cell migration. Science. 2016;352:353–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Raab M, Gentili M, de Belly H, Thiam HR, Vargas P, Jimenez AJ, et al. ESCRT III repairs nuclear envelope ruptures during cell migration to limit DNA damage and cell death. Science. 2016;352:359–62.

    Article  CAS  PubMed  Google Scholar 

  32. Mitchell MJ, Denais C, Chan MF, Wang Z, Lammerding J, King MR. Lamin A/C deficiency reduces circulating tumor cell resistance to fluid shear stress. Am J Physiol Cell Physiol. 2015;309:C736–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gruenbaum Y, Foisner R. Lamins: nuclear intermediate filament proteins with fundamental functions in nuclear mechanics and genome regulation. Annu Rev Biochem. 2015;84:131–64.

    Article  CAS  PubMed  Google Scholar 

  34. Andres V, Gonzalez JM. Role of A-type lamins in signaling, transcription, and chromatin organization. J Cell Biol. 2009;187:945–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Maurer M, Lammerding J. The driving force: nuclear mechanotransduction in cellular function, fate, and disease. Annu Rev Biomed Eng. 2019;21:443–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Schreiber KH, Kennedy BK. When lamins go bad: nuclear structure and disease. Cell. 2013;152:1365–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wong X, Stewart CL. The laminopathies and the insights they provide into the structural and functional organization of the nucleus. Annu Rev Genomics Hum Genet. 2020;21:263–88.

    Article  CAS  PubMed  Google Scholar 

  38. Dubik N, Mai S. Lamin A/C: function in normal and tumor cells. Cancers. 2020;12:3688.

    Article  CAS  PubMed Central  Google Scholar 

  39. Alhudiri IM, Nolan CC, Ellis IO, Elzagheid A, Rakha EA, Green AR, et al. Expression of Lamin A/C in early-stage breast cancer and its prognostic value. Breast Cancer Res Treat. 2019;174:661–8.

    Article  CAS  PubMed  Google Scholar 

  40. Matsumoto A, Hieda M, Yokoyama Y, Nishioka Y, Yoshidome K, Tsujimoto M, et al. Global loss of a nuclear lamina component, lamin A/C, and LINC complex components SUN1, SUN2, and nesprin-2 in breast cancer. Cancer Med. 2015;4:1547–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wazir U, Ahmed MH, Bridger JM, Harvey A, Jiang WG, Sharma AK, et al. The clinicopathological significance of lamin A/C, lamin B1 and lamin B receptor mRNA expression in human breast cancer. Cell Mol Biol Lett. 2013;18:595–611.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Capo-chichi CD, Cai KQ, Smedberg J, Ganjei-Azar P, Godwin AK, Xu XX. Loss of A-type lamin expression compromises nuclear envelope integrity in breast cancer. Chin J Cancer. 2011;30:415–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Onitilo AA, Engel JM, Greenlee RT, Mukesh BN. Breast cancer subtypes based on ER/PR and Her2 expression: comparison of clinicopathologic features and survival. Clin Med Res. 2009;7:4–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA. 2001;98:10869–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Davidson PM, Fedorchak GR, Mondesert-Deveraux S, Bell ES, Isermann P, Aubry D, et al. High-throughput microfluidic micropipette aspiration device to probe time-scale dependent nuclear mechanics in intact cells. Lab Chip. 2019;19:3652–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Saleh T, Alhesa A, El-Sadoni M, Abu Shahin N, Alsharaiah E, Al Shboul S, et al. The expression of the senescence-associated biomarker Lamin B1 in human breast cancer. Diagnostics. 2020;12:609.

    Article  CAS  Google Scholar 

  47. Stewart C, Burke B. Teratocarcinoma stem cells and early mouse embryos contain only a single major lamin polypeptide closely resembling lamin B. Cell. 1987;51:383–92.

    Article  CAS  PubMed  Google Scholar 

  48. Bussolati G, Maletta F, Asioli S, Annaratone L, Sapino A, Marchio C. “To be or not to be in a good shape”: diagnostic and clinical value of nuclear shape irregularities in thyroid and breast cancer. Adv Exp Med Biol. 2014;773:101–21.

    Article  CAS  PubMed  Google Scholar 

  49. Pajerowski JD, Dahl KN, Zhong FL, Sammak PJ, Discher DE. Physical plasticity of the nucleus in stem cell differentiation. Proc Natl Acad Sci USA. 2007;104:15619–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wang X, Liu H, Zhu M, Cao C, Xu Z, Tsatskis Y, et al. Mechanical stability of the cell nucleus—roles played by the cytoskeleton in nuclear deformation and strain recovery. J Cell Sci. 2018;131:jcs209627.

    Article  PubMed  CAS  Google Scholar 

  51. Lee H, Adams WJ, Alford PW, McCain ML, Feinberg AW, Sheehy SP, et al. Cytoskeletal prestress regulates nuclear shape and stiffness in cardiac myocytes. Exp Biol Med. 2015;240:1543–54.

    Article  CAS  Google Scholar 

  52. McGregor AL, Hsia CR, Lammerding J. Squish and squeeze-the nucleus as a physical barrier during migration in confined environments. Curr Opin Cell Biol. 2016;40:32–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Davidson PM, Sliz J, Isermann P, Denais C, Lammerding J. Design of a microfluidic device to quantify dynamic intra-nuclear deformation during cell migration through confining environments. Integr Biol. 2015;7:1534–46.

    Article  CAS  Google Scholar 

  54. Guy CT, Cardiff RD, Muller WJ. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol Cell Biol. 1992;12:954–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Borowsky AD, Namba R, Young LJ, Hunter KW, Hodgson JG, Tepper CG, et al. Syngeneic mouse mammary carcinoma cell lines: two closely related cell lines with divergent metastatic behavior. Clin Exp Metastasis. 2005;22:47–59.

    Article  CAS  PubMed  Google Scholar 

  56. Mekhdjian AH, Kai F, Rubashkin MG, Prahl LS, Przybyla LM, McGregor AL, et al. Integrin-mediated traction force enhances paxillin molecular associations and adhesion dynamics that increase the invasiveness of tumor cells into a three-dimensional extracellular matrix. Mol Biol Cell. 2017;28:1467–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sahai E. Illuminating the metastatic process. Nat Rev Cancer. 2007;7:737–49.

    Article  CAS  PubMed  Google Scholar 

  58. Aslakson CJ, Miller FR. Selective events in the metastatic process defined by analysis of the sequential dissemination of subpopulations of a mouse mammary tumor. Cancer Res. 1992;52:1399–405.

    CAS  PubMed  Google Scholar 

  59. Dexter DL, Kowalski HM, Blazar BA, Fligiel Z, Vogel R, Heppner GH. Heterogeneity of tumor cells from a single mouse mammary tumor. Cancer Res. 1978;38:3174–81.

    CAS  PubMed  Google Scholar 

  60. Lelekakis M, Moseley JM, Martin TJ, Hards D, Williams E, Ho P, et al. A novel orthotopic model of breast cancer metastasis to bone. Clin Exp Metastasis. 1999;17:163–70.

    Article  CAS  PubMed  Google Scholar 

  61. de Leeuw R, Gruenbaum Y, Medalia O. Nuclear lamins: thin filaments with major functions. Trends Cell Biol. 2018;28:34–45.

    Article  PubMed  CAS  Google Scholar 

  62. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47:D607–13.

    Article  CAS  PubMed  Google Scholar 

  63. Bertacchini J, Beretti F, Cenni V, Guida M, Gibellini F, Mediani L, et al. The protein kinase Akt/PKB regulates both prelamin A degradation and Lmna gene expression. FASEB J. 2013;27:2145–55.

    Article  CAS  PubMed  Google Scholar 

  64. Lloyd DJ, Trembath RC, Shackleton S. A novel interaction between lamin A and SREBP1: implications for partial lipodystrophy and other laminopathies. Hum Mol Genet. 2002;11:769–77.

    Article  CAS  PubMed  Google Scholar 

  65. Okumura K, Hosoe Y, Nakajima N. c-Jun and Sp1 family are critical for retinoic acid induction of the lamin A/C retinoic acid-responsive element. Biochem Biophys Res Commun. 2004;320:487–92.

    Article  CAS  PubMed  Google Scholar 

  66. Mymrikov EV, Seit-Nebi AS, Gusev NB. Large potentials of small heat shock proteins. Physiol Rev. 2011;91:1123–59.

    Article  CAS  PubMed  Google Scholar 

  67. Batulan Z, Pulakazhi Venu VK, Li Y, Koumbadinga G, Alvarez-Olmedo DG, Shi C, et al. Extracellular release and signaling by heat shock protein 27: role in modifying vascular inflammation. Front Immunol. 2016;7:285.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Sugiyama Y, Suzuki A, Kishikawa M, Akutsu R, Hirose T, Waye MM, et al. Muscle develops a specific form of small heat shock protein complex composed of MKBP/HSPB2 and HSPB3 during myogenic differentiation. J Biol Chem. 2000;275:1095–104.

    Article  CAS  PubMed  Google Scholar 

  69. Isermann P, Lammerding J. Nuclear mechanics and mechanotransduction in health and disease. Curr Biol. 2013;23:R1113–21.

    Article  CAS  PubMed  Google Scholar 

  70. Buxboim A, Swift J, Irianto J, Spinler KR, Dingal PC, Athirasala A, et al. Matrix elasticity regulates lamin-A,C phosphorylation and turnover with feedback to actomyosin. Curr Biol. 2014;24:1909–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Peter M, Nakagawa J, Doree M, Labbe JC, Nigg EA. In vitro disassembly of the nuclear lamina and M phase-specific phosphorylation of lamins by cdc2 kinase. Cell. 1990;61:591–602.

    Article  CAS  PubMed  Google Scholar 

  72. Collas P. Sequential PKC- and Cdc2-mediated phosphorylation events elicit zebrafish nuclear envelope disassembly. J Cell Sci. 1999;112:977–87.

    Article  CAS  PubMed  Google Scholar 

  73. Barati MT, Rane MJ, Klein JB, McLeish KR. A proteomic screen identified stress-induced chaperone proteins as targets of Akt phosphorylation in mesangial cells. J Proteome Res. 2006;5:1636–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Cenni V, Bertacchini J, Beretti F, Lattanzi G, Bavelloni A, Riccio M, et al. Lamin A Ser404 is a nuclear target of Akt phosphorylation in C2C12 cells. J Proteome Res. 2008;7:4727–35.

    Article  CAS  PubMed  Google Scholar 

  75. Naeem AS, Zhu Y, Di WL, Marmiroli S, O’Shaughnessy RF. AKT1-mediated lamin A/C degradation is required for nuclear degradation and normal epidermal terminal differentiation. Cell Death Differ. 2015;22:2123–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Fruman DA, Rommel C. PI3K and cancer: lessons, challenges and opportunities. Nat Rev Drug Discov. 2014;13:140–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Wennemers M, Stegeman H, Bussink J, Versleijen-Jonkers YM, van Laarhoven HW, Raleigh JA, et al. Hypoxia regulation of phosphokinases and the prognostic value of pAKT in breast cancer. Int J Biol Markers. 2013;28:151–60.

    Article  CAS  PubMed  Google Scholar 

  78. Rhodes N, Heerding DA, Duckett DR, Eberwein DJ, Knick VB, Lansing TJ, et al. Characterization of an Akt kinase inhibitor with potent pharmacodynamic and antitumor activity. Cancer Res. 2008;68:2366–74.

    Article  CAS  PubMed  Google Scholar 

  79. Han EK, Leverson JD, McGonigal T, Shah OJ, Woods KW, Hunter T, et al. Akt inhibitor A-443654 induces rapid Akt Ser-473 phosphorylation independent of mTORC1 inhibition. Oncogene. 2007;26:5655–61.

    Article  CAS  PubMed  Google Scholar 

  80. Maira SM, Pecchi S, Huang A, Burger M, Knapp M, Sterker D, et al. Identification and characterization of NVP-BKM120, an orally available pan-class I PI3-kinase inhibitor. Mol Cancer Ther. 2012;11:317–28.

    Article  CAS  PubMed  Google Scholar 

  81. Agrelo R, Setien F, Espada J, Artiga MJ, Rodriguez M, Perez-Rosado A, et al. Inactivation of the lamin A/C gene by CpG island promoter hypermethylation in hematologic malignancies, and its association with poor survival in nodal diffuse large B-cell lymphoma. J Clin Oncol. 2005;23:3940–7.

    Article  CAS  PubMed  Google Scholar 

  82. Marmiroli S, Bertacchini J, Beretti F, Cenni V, Guida M, De Pol A, et al. A-type lamins and signaling: the PI 3-kinase/Akt pathway moves forward. J Cell Physiol. 2009;220:553–61.

    Article  CAS  PubMed  Google Scholar 

  83. Nagelkerke A, van Kuijk SJ, Sweep FC, Nagtegaal ID, Hoogerbrugge N, Martens JW, et al. Constitutive expression of gamma-H2AX has prognostic relevance in triple negative breast cancer. Radiother Oncol. 2011;101:39–45.

    Article  CAS  PubMed  Google Scholar 

  84. Cho S, Irianto J, Discher DE. Mechanosensing by the nucleus: From pathways to scaling relationships. J Cell Biol. 2017;216:305–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Gonzalez-Cruz RD, Sadick JS, Fonseca VC, Darling EM. Nuclear lamin protein C is linked to lineage-specific, whole-cell mechanical properties. Cell Mol Bioeng. 2018;11:131–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Ortega MA, Fraile-Martinez O, Asunsolo A, Bujan J, Garcia-Honduvilla N, Coca S. Signal transduction pathways in breast cancer: the important role of PI3K/Akt/mTOR. J Oncol. 2020;2020:9258396.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Broers JL, Machiels BM, Kuijpers HJ, Smedts F, van den Kieboom R, Raymond Y, et al. A- and B-type lamins are differentially expressed in normal human tissues. Histochem Cell Biol. 1997;107:505–17.

    Article  CAS  PubMed  Google Scholar 

  88. Ivorra C, Kubicek M, Gonzalez JM, Sanz-Gonzalez SM, Alvarez-Barrientos A, O’Connor JE, et al. A mechanism of AP-1 suppression through interaction of c-Fos with lamin A/C. Genes Dev. 2006;20:307–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Nitta RT, Jameson SA, Kudlow BA, Conlan LA, Kennedy BK. Stabilization of the retinoblastoma protein by A-type nuclear lamins is required for INK4A-mediated cell cycle arrest. Mol Cell Biol. 2006;26:5360–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Van Berlo JH, Voncken JW, Kubben N, Broers JL, Duisters R, van Leeuwen RE, et al. A-type lamins are essential for TGF-beta1 induced PP2A to dephosphorylate transcription factors. Hum Mol Genet. 2005;14:2839–49.

    Article  PubMed  Google Scholar 

  91. Johnson BR, Nitta RT, Frock RL, Mounkes L, Barbie DA, Stewart CL, et al. A-type lamins regulate retinoblastoma protein function by promoting subnuclear localization and preventing proteasomal degradation. Proc Natl Acad Sci USA. 2004;101:9677–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Aljada A, Doria J, Saleh AM, Al-Matar SH, AlGabbani S, Shamsa HB, et al. Altered Lamin A/C splice variant expression as a possible diagnostic marker in breast cancer. Cell Oncol. 2016;39:161–74.

    Article  CAS  Google Scholar 

  93. Belt EJ, Fijneman RJ, van den Berg EG, Bril H, Delis-van Diemen PM, Tijssen M, et al. Loss of lamin A/C expression in stage II and III colon cancer is associated with disease recurrence. Eur J Cancer. 2011;47:1837–45.

    Article  CAS  PubMed  Google Scholar 

  94. Wu Z, Wu L, Weng D, Xu D, Geng J, Zhao F. Reduced expression of lamin A/C correlates with poor histological differentiation and prognosis in primary gastric carcinoma. J Exp Clin Cancer Res. 2009;28:8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Coradeghini R, Barboro P, Rubagotti A, Boccardo F, Parodi S, Carmignani G, et al. Differential expression of nuclear lamins in normal and cancerous prostate tissues. Oncol Rep. 2006;15:609–13.

    CAS  PubMed  Google Scholar 

  96. Willis ND, Cox TR, Rahman-Casans SF, Smits K, Przyborski SA, van den Brandt P, et al. Lamin A/C is a risk biomarker in colorectal cancer. PLoS ONE. 2008;3:e2988.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Gong G, Chen P, Li L, Tan H, Zhou J, Zhou Y, et al. Loss of lamin A but not lamin C expression in epithelial ovarian cancer cells is associated with metastasis and poor prognosis. Pathol Res Pract. 2015;211:175–82.

    Article  CAS  PubMed  Google Scholar 

  98. Venables RS, McLean S, Luny D, Moteleb E, Morley S, Quinlan RA, et al. Expression of individual lamins in basal cell carcinomas of the skin. Br J Cancer. 2001;84:512–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Shimi T, Pfleghaar K, Kojima S, Pack CG, Solovei I, Goldman AE, et al. The A- and B-type nuclear lamin networks: microdomains involved in chromatin organization and transcription. Genes Dev. 2008;22:3409–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Fong LG, Ng JK, Lammerding J, Vickers TA, Meta M, Cote N, et al. Prelamin A and lamin A appear to be dispensable in the nuclear lamina. J Clin Investig. 2006;116:743–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Sullivan T, Escalante-Alcalde D, Bhatt H, Anver M, Bhat N, Nagashima K, et al. Loss of A-type lamin expression compromises nuclear envelope integrity leading to muscular dystrophy. J Cell Biol. 1999;147:913–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Kennedy BK, Pennypacker JK. RB and lamins in cell cycle regulation and aging. Adv Exp Med Biol. 2014;773:127–42.

    Article  CAS  PubMed  Google Scholar 

  103. Redwood AB, Perkins SM, Vanderwaal RP, Feng Z, Biehl KJ, Gonzalez-Suarez I, et al. A dual role for A-type lamins in DNA double-strand break repair. Cell Cycle. 2011;10:2549–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Maynard S, Keijzers G, Akbari M, Ezra MB, Hall A, Morevati M, et al. Lamin A/C promotes DNA base excision repair. Nucleic Acids Res. 2019;47:11709–28.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Gonzalo S. DNA damage and lamins. Adv Exp Med Biol. 2014;773:377–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Frock RL, Kudlow BA, Evans AM, Jameson SA, Hauschka SD, Kennedy BK. Lamin A/C and emerin are critical for skeletal muscle satellite cell differentiation. Genes Dev. 2006;20:486–500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Hoxhaj G, Manning BD. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat Rev Cancer. 2020;20:74–88.

    Article  CAS  PubMed  Google Scholar 

  108. Ali R, Wendt MK. The paradoxical functions of EGFR during breast cancer progression. Signal Transduct Target Ther. 2017;2:16042.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Brady CA, Attardi LD. p53 at a glance. J Cell Sci. 2010;123:2527–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Concin N, Zeillinger C, Tong D, Stimpfl M, Konig M, Printz D, et al. Comparison of p53 mutational status with mRNA and protein expression in a panel of 24 human breast carcinoma cell lines. Breast Cancer Res Treat. 2003;79:37–46.

    Article  CAS  PubMed  Google Scholar 

  111. Condor M, Mark C, Gerum RC, Grummel NC, Bauer A, Garcia-Aznar JM, et al. Breast cancer cells adapt contractile forces to overcome steric hindrance. Biophys J. 2019;116:1305–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Shah P, Hobson CM, Cheng S, Colville MJ, Paszek MJ, Superfine R, et al. Nuclear deformation causes DNA damage by increasing replication stress. Curr Biol. 2021;31:753–65.e6.

    Article  CAS  PubMed  Google Scholar 

  113. Irianto J, Pfeifer CR, Bennett RR, Xia Y, Ivanovska IL, Liu AJ, et al. Nuclear constriction segregates mobile nuclear proteins away from chromatin. Mol Biol Cell. 2016;27:4011–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Roncato F, Regev O, Feigelson SW, Yadav SK, Kaczmarczyk L, Levi N, et al. Reduced lamin A/C does not facilitate cancer cell transendothelial migration but compromises lung metastasis. Cancers. 2021;13:2383.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Pickup MW, Laklai H, Acerbi I, Owens P, Gorska AE, Chytil A, et al. Stromally derived lysyl oxidase promotes metastasis of transforming growth factor-beta-deficient mouse mammary carcinomas. Cancer Res. 2013;73:5336–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Miroshnikova YA, Mouw JK, Barnes JM, Pickup MW, Lakins JN, Kim Y, et al. Tissue mechanics promote IDH1-dependent HIF1alpha-tenascin C feedback to regulate glioblastoma aggression. Nat Cell Biol. 2016;18:1336–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Hanson PI, Roth R, Lin Y, Heuser JE. Plasma membrane deformation by circular arrays of ESCRT-III protein filaments. J Cell Biol. 2008;180:389–402.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Guz N, Dokukin M, Kalaparthi V, Sokolov I. If cell mechanics can be described by elastic modulus: study of different models and probes used in indentation experiments. Biophys J. 2014;107:564–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, et al. Fiji: an open-source platform for biological-image analysis. Nat Methods. 2012;9:676–82.

    Article  CAS  PubMed  Google Scholar 

  120. Bastos de Oliveira FM, Kim D, Cussiol JR, Das J, Jeong MC, Doerfler L, et al. Phosphoproteomics reveals distinct modes of Mec1/ATR signaling during DNA replication. Mol Cell. 2015;57:1124–32.

    Article  CAS  PubMed  Google Scholar 

  121. Bastos de Oliveira FM, Kim D, Lanz M, Smolka MB. Quantitative analysis of DNA damage signaling responses to chemical and genetic perturbations. Methods Mol Biol. 2018;1672:645–60.

    Article  CAS  PubMed  Google Scholar 

  122. Kim D, Liu Y, Oberly S, Freire R, Smolka MB. ATR-mediated proteome remodeling is a major determinant of homologous recombination capacity in cancer cells. Nucleic Acids Res. 2018;46:8311–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. R_Core_Team. R: A language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing; 2021. https://www.R-project.org/.

  124. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Kassambara A. ggpubr: ‘ggplot2’ Based Publication Ready Plots. R package version 040. 2020. https://CRAN.R-project.org/package=ggpubr.

  126. Kramer A, Green J, Pollard J Jr., Tugendreich S. Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics. 2014;30:523–30.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank Prof. Peter Friedl for the 4T1 progression series cell lines. This work was performed in part at the Cornell NanoScale Science & Technology Facility, a member of the National Nanotechnology Coordinated Infrastructure, which is supported by the National Science Foundation (Grant NNCI-2025233). The content of this paper is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. The results published here are in part based upon data generated by the TCGA Research Network: https://www.cancer.gov/tcga.

Funding

This work was supported by funding from the National Institutes of Health (R01 HL082792, R01 GM137605, U54 CA210184, and U54 CA193461 to JL, R35 GM141159 and R01 GM123018 to MBS), the Department of Defense Breast Cancer Research Program (Breakthrough Award BC150580 to JL), and the National Science Foundation (CAREER Award CBET-1254846 to JL and Graduate Research Fellowship DGE-1144153 to ALM). This work was performed in part at the Cornell NanoScale Science & Technology Facility (CNF), a member of the National Nanotechnology Coordinated Infrastructure NNCI), which is supported by the National Science Foundation (Grant NNCI-2025233).

Author information

Authors and Affiliations

Authors

Contributions

ESB and JL conceptualized and designed the experiments; JL supervised the research; ESB, PS, NZS, AAV, JLPM, and ALM performed experiments and analyzed data; DK and MS performed the SILAC proteomic analysis; PI, JJE, PMD, JNL, and VMW contributed to the development of resources, including constructs, cell lines, assays, and/or image analysis methods; PNS and LV contributed human breast tumor tissue samples and analysis; EB and JL wrote the paper; all authors contributed to the editing of the paper; and JL and MBS acquired funding.

Corresponding author

Correspondence to Jan Lammerding.

Ethics declarations

Competing interests

JL has provided paid consulting services for BridgeBio for the role of lamins in unrelated projects.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bell, E.S., Shah, P., Zuela-Sopilniak, N. et al. Low lamin A levels enhance confined cell migration and metastatic capacity in breast cancer. Oncogene 41, 4211–4230 (2022). https://doi.org/10.1038/s41388-022-02420-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-022-02420-9

This article is cited by

Search

Quick links