Skip to main content
Log in

Nuclear Deformability Constitutes a Rate-Limiting Step During Cell Migration in 3-D Environments

  • Published:
Cellular and Molecular Bioengineering Aims and scope Submit manuscript

Abstract

Cell motility plays a critical role in many physiological and pathological settings, ranging from wound healing to cancer metastasis. While cell migration on 2-dimensional (2-D) substrates has been studied for decades, the physical challenges cells face when moving in 3-D environments are only now emerging. In particular, the cell nucleus, which occupies a large fraction of the cell volume and is normally substantially stiffer than the surrounding cytoplasm, may impose a major obstacle when cells encounter narrow constrictions in the interstitial space, the extracellular matrix, or small capillaries. Using novel microfluidic devices that allow observation of cells moving through precisely defined geometries at high spatial and temporal resolution, we determined nuclear deformability as a critical factor in the cells’ ability to pass through constrictions smaller than the size of the nucleus. Furthermore, we found that cells with reduced levels of the nuclear envelope proteins lamins A/C, which are the main determinants of nuclear stiffness, passed significantly faster through narrow constrictions during active migration and passive perfusion. Given recent reports that many human cancers have altered lamin expression, our findings suggest a novel biophysical mechanism by which changes in nuclear structure and composition may promote cancer cell invasion and metastasis.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6
Figure 7

Similar content being viewed by others

Abbreviations

2-D:

Two-dimensional

3-D:

Three-dimensional

ANOVA:

Analysis of variance

BSA:

Bovine serum albumin

DMEM:

Dulbecco Modified Eagle Medium

FBS:

Fetal bovine serum

GFP:

Green fluorescent protein

LINC:

Linker of nucleoskeleton and cytoskeleton

MEF:

Mouse embryonic fibroblast

MMP:

Matrix metalloproteinase

PBS:

Phosphate buffered saline

PDGF:

Platelet derived growth factor

PDMS:

Polydimethylsiloxane

References

  1. Balzer, E. M., Z. Tong, C. D. Paul, W. C. Hung, K. M. Stroka, A. E. Boggs, S. S. Martin, and K. Konstantopoulos. Physical confinement alters tumor cell adhesion and migration phenotypes. FASEB J. 26(10):4045–4056, 2012.

    Article  Google Scholar 

  2. Booth-Gauthier, E. A., V. Du, M. Ghibaudo, A. D. Rape, K. N. Dahl, and B. Ladoux. Hutchinson–Gilford progeria syndrome alters nuclear shape and reduces cell motility in three dimensional model substrates. Integr. Biol. (Camb.) 5(3):569–577, 2013.

    Article  Google Scholar 

  3. Capo-chichi, C. D., K. Q. Cai, J. Smedberg, P. Ganjei-Azar, A. K. Godwin, and X. X. Xu. Loss of A-type lamin expression compromises nuclear envelope integrity in breast cancer. Chin. J. Cancer 30(6):415–425, 2011.

    Article  Google Scholar 

  4. Chaffer, C. L., and R. A. Weinberg. A perspective on cancer cell metastasis. Science 331(6024):1559–1564, 2011.

    Article  Google Scholar 

  5. Crisp, M., Q. Liu, K. Roux, J. B. Rattner, C. Shanahan, B. Burke, P. D. Stahl, and D. Hodzic. Coupling of the nucleus and cytoplasm: role of the LINC complex. J. Cell Biol. 172(1):41–53, 2006.

    Article  Google Scholar 

  6. Dahl, K. N., P. Scaffidi, M. F. Islam, A. G. Yodh, K. L. Wilson, and T. Misteli. Distinct structural and mechanical properties of the nuclear lamina in Hutchinson–Gilford progeria syndrome. Proc. Natl. Acad. Sci. USA 103(27):10271–10276, 2006.

    Article  Google Scholar 

  7. Davidson, P. M., and J. Lammerding. Broken nuclei—lamins, nuclear mechanics, and disease. Trends Cell Biol. 24(4):247–256, 2014.

  8. Deguchi, S., K. Maeda, T. Ohashi, and M. Sato. Flow-induced hardening of endothelial nucleus as an intracellular stress-bearing organelle. J. Biomech. 38(9):1751–1759, 2005.

    Article  Google Scholar 

  9. Fields, A. P., G. Tyler, A. S. Kraft, and W. S. May. Role of nuclear protein kinase C in the mitogenic response to platelet-derived growth factor. J. Cell Sci. 96(Pt 1):107–114, 1990.

    Google Scholar 

  10. Folker, E. S., C. Ostlund, G. W. Luxton, H. J. Worman, and G. G. Gundersen. Lamin A variants that cause striated muscle disease are defective in anchoring transmembrane actin-associated nuclear lines for nuclear movement. Proc. Natl. Acad. Sci. USA 108(1):131–136, 2011.

    Article  Google Scholar 

  11. Friedl, P., E. Sahai, S. Weiss, and K. M. Yamada. New dimensions in cell migration. Nat. Rev. Mol. Cell Biol. 13(11):743–747, 2012.

    Article  Google Scholar 

  12. Friedl, P., and K. Wolf. Plasticity of cell migration: a multiscale tuning model. J. Cell Biol. 188(1):11–19, 2010.

    Article  Google Scholar 

  13. Friedl, P., K. Wolf, and J. Lammerding. Nuclear mechanics during cell migration. Curr. Opin. Cell Biol. 23(1):55–64, 2011.

    Article  Google Scholar 

  14. Fu, Y., L. K. Chin, T. Bourouina, A. Q. Liu, and A. M. VanDongen. Nuclear deformation during breast cancer cell transmigration. Lab Chip 12(19):3774–3778, 2012.

    Article  Google Scholar 

  15. Glynn, M. W., and T. W. Glover. Incomplete processing of mutant lamin A in Hutchinson–Gilford progeria leads to nuclear abnormalities, which are reversed by farnesyltransferase inhibition. Hum. Mol. Genet. 14(20):2959–2969, 2005.

    Article  Google Scholar 

  16. Gundersen, G. G., and H. J. Worman. Nuclear positioning. Cell 152(6):1376–1389, 2013.

    Article  Google Scholar 

  17. Hale, C. M., A. L. Shrestha, S. B. Khatau, P. J. Stewart-Hutchinson, L. Hernandez, C. L. Stewart, D. Hodzic, and D. Wirtz. Dysfunctional connections between the nucleus and the actin and microtubule networks in laminopathic models. Biophys. J. 95(11):5462–5475, 2008.

    Article  Google Scholar 

  18. Harada, T., J. Swift, J. Irianto, J. W. Shin, K. R. Spinler, A. Athirasala, R. Diegmiller, P. C. Dingal, I. L. Ivanovska, and D. E. Discher. Nuclear lamin stiffness is a barrier to 3D migration, but softness can limit survival. J. Cell Biol. 204(5):669–682, 2014.

    Article  Google Scholar 

  19. Ho, C. Y., and J. Lammerding. Lamins at a glance. J. Cell Sci. 125(Pt 9):2087–2093, 2012.

    Article  Google Scholar 

  20. Hung, W. C., S. H. Chen, C. D. Paul, K. M. Stroka, Y. C. Lo, J. T. Yang, and K. Konstantopoulos. Distinct signaling mechanisms regulate migration in unconfined vs. confined spaces. J. Cell Biol. 202(5):807–824, 2013.

    Article  Google Scholar 

  21. Isermann, P., P. M. Davidson, J. D. Sliz, and J. Lammerding. Assays to measure nuclear mechanics in interphase cells. Curr. Protoc. Cell Biol. Chapter 22:Unit22.16, 2012. doi:10.1002/0471143030.cb2216s56.

  22. Ivanovska, I., J. Swift, T. Harada, J. D. Pajerowski, and D. E. Discher. Physical plasticity of the nucleus and its manipulation. Methods Cell Biol. 98:207–220, 2010.

    Article  Google Scholar 

  23. Khatau, S. B., R. J. Bloom, S. Bajpai, D. Razafsky, S. Zang, A. Giri, P. H. Wu, J. Marchand, A. Celedon, C. M. Hale, S. X. Sun, D. Hodzic, and D. Wirtz. The distinct roles of the nucleus and nucleus-cytoskeleton connections in three-dimensional cell migration. Sci. Rep. 2:488, 2012.

    Article  Google Scholar 

  24. Lammerding, J., L. G. Fong, J. Y. Ji, K. Reue, C. L. Stewart, S. G. Young, and R. T. Lee. Lamins A and C but not lamin B1 regulate nuclear mechanics. J. Biol. Chem. 281(35):25768–25780, 2006.

    Article  Google Scholar 

  25. Lammerding, J., P. C. Schulze, T. Takahashi, S. Kozlov, T. Sullivan, R. D. Kamm, C. L. Stewart, and R. T. Lee. Lamin A/C deficiency causes defective nuclear mechanics and mechanotransduction. J. Clin. Investig. 113(3):370–378, 2004.

    Article  Google Scholar 

  26. Lee, J. S., C. M. Hale, P. Panorchan, S. B. Khatau, J. P. George, Y. Tseng, C. L. Stewart, D. Hodzic, and D. Wirtz. Nuclear lamin A/C deficiency induces defects in cell mechanics, polarization, and migration. Biophys. J. 93(7):2542–2552, 2007.

    Article  Google Scholar 

  27. Lombardi, M. L., D. E. Jaalouk, C. M. Shanahan, B. Burke, K. J. Roux, and J. Lammerding. The interaction between nesprins and sun proteins at the nuclear envelope is critical for force transmission between the nucleus and cytoskeleton. J. Biol. Chem. 286(30):26743–26753, 2011.

    Article  Google Scholar 

  28. Luxton, G. W., E. R. Gomes, E. S. Folker, E. Vintinner, and G. G. Gundersen. Linear arrays of nuclear envelope proteins harness retrograde actin flow for nuclear movement. Science 329(5994):956–959, 2010.

    Article  Google Scholar 

  29. Olins, A. L., T. V. Hoang, M. Zwerger, H. Herrmann, H. Zentgraf, A. A. Noegel, I. Karakesisoglou, D. Hodzic, and D. E. Olins. The LINC-less granulocyte nucleus. Eur. J. Cell Biol. 88(4):203–214, 2009.

    Article  Google Scholar 

  30. Pajerowski, J. D., K. N. Dahl, F. L. Zhong, P. J. Sammak, and D. E. Discher. Physical plasticity of the nucleus in stem cell differentiation. Proc. Natl. Acad. Sci. USA 104(40):15619–15624, 2007.

    Article  Google Scholar 

  31. Petrie, R. J., and K. M. Yamada. At the leading edge of three-dimensional cell migration. J. Cell Sci. 125(Pt 24):5917–5926, 2012.

    Article  Google Scholar 

  32. Rosenbluth, M. J., W. A. Lam, and D. A. Fletcher. Analyzing cell mechanics in hematologic diseases with microfluidic biophysical flow cytometry. Lab Chip 8(7):1062–1070, 2008.

    Article  Google Scholar 

  33. Rowat, A. C., D. E. Jaalouk, M. Zwerger, W. L. Ung, I. A. Eydelnant, D. E. Olins, A. L. Olins, H. Herrmann, D. A. Weitz, and J. Lammerding. Nuclear envelope composition determines the ability of neutrophil-type cells to passage through micron-scale constrictions. J. Biol. Chem. 288(12):8610–8618, 2013.

    Article  Google Scholar 

  34. Shao, J. Y., and J. Xu. A modified micropipette aspiration technique and its application to tether formation from human neutrophils. J. Biomech. Eng. 124(4):388–396, 2002.

    Article  MathSciNet  Google Scholar 

  35. Shin, J. W., K. R. Spinler, J. Swift, J. A. Chasis, N. Mohandas, and D. E. Discher. Lamins regulate cell trafficking and lineage maturation of adult human hematopoietic cells. Proc. Natl. Acad. Sci. USA 110(47):18892–18897, 2013.

    Article  Google Scholar 

  36. Stoitzner, P., K. Pfaller, H. Stossel, and N. Romani. A close-up view of migrating Langerhans cells in the skin. J. Investig. Dermatol. 118(1):117–125, 2002.

    Article  Google Scholar 

  37. Sullivan, T., D. Escalante-Alcalde, H. Bhatt, M. Anver, N. Bhat, K. Nagashima, C. L. Stewart, and B. Burke. Loss of A-type lamin expression compromises nuclear envelope integrity leading to muscular dystrophy. J. Cell Biol. 147(5):913–920, 1999.

    Article  Google Scholar 

  38. Swift, J., I. L. Ivanovska, A. Buxboim, T. Harada, P. C. Dingal, J. Pinter, J. D. Pajerowski, K. R. Spinler, J. W. Shin, M. Tewari, F. Rehfeldt, D. W. Speicher, and D. E. Discher. Nuclear lamin-A scales with tissue stiffness and enhances matrix-directed differentiation. Science 341(6149):1240104, 2013.

    Article  Google Scholar 

  39. Tong, Z., E. M. Balzer, M. R. Dallas, W. C. Hung, K. J. Stebe, and K. Konstantopoulos. Chemotaxis of cell populations through confined spaces at single-cell resolution. PLoS ONE 7(1):e29211, 2012.

    Article  Google Scholar 

  40. Verstraeten, V. L., J. Y. Ji, K. S. Cummings, R. T. Lee, and J. Lammerding. Increased mechanosensitivity and nuclear stiffness in Hutchinson–Gilford progeria cells: effects of farnesyltransferase inhibitors. Aging Cell 7(3):383–393, 2008.

    Article  Google Scholar 

  41. Verstraeten, V. L., L. A. Peckham, M. Olive, B. C. Capell, F. S. Collins, E. G. Nabel, S. G. Young, L. G. Fong, and J. Lammerding. Protein farnesylation inhibitors cause donut-shaped cell nuclei attributable to a centrosome separation defect. Proc. Natl. Acad. Sci. USA 108(12):4997–5002, 2011.

    Article  Google Scholar 

  42. Wazir, U., M. H. Ahmed, J. M. Bridger, A. Harvey, W. G. Jiang, A. K. Sharma, and K. Mokbel. The clinicopathological significance of lamin A/C, lamin B1 and lamin B receptor mRNA expression in human breast cancer. Cell. Mol. Biol. Lett. 18(4):595–611, 2013.

    Article  Google Scholar 

  43. Weigelin, B., G.-J. Bakker, and P. Friedl. Intravital third harmonic generation microscopy of collective melanoma cell invasion. Principles of interface guidance and microvesicle dynamics. IntraVital 1(1):32–43, 2012.

    Article  Google Scholar 

  44. Wolf, K., M. Te Lindert, M. Krause, S. Alexander, J. Te Riet, A. L. Willis, R. M. Hoffman, C. G. Figdor, S. J. Weiss, and P. Friedl. Physical limits of cell migration: control by ECM space and nuclear deformation and tuning by proteolysis and traction force. J. Cell Biol. 201(7):1069–1084, 2013.

    Article  Google Scholar 

Download references

Acknowledgments

The authors thank Philipp Isermann for providing plasmids (GFP-LifeAct, mCherry–Histone-4), advice, and some MATLAB image analysis scripts; Drs. Colin Stewart and Tom Glover for providing cells and reagents; Dr. Amy Rowat for design and fabrication of some of the perfusion devices; Dr. Sarah Jandricic for help with statistical analysis; Kathy Zhang and Ileana D’Aloisio for quantification of cell migration assays; and Rachel Gilbert for helpful discussions. This work was performed in part at the Cornell NanoScale Facility, a member of the National Nanotechnology Infrastructure Network, which is supported by the National Science Foundation (Grant ECCS-0335765). This work was supported by National Institutes of Health awards [R01 NS059348 and R01 HL082792]; the Department of Defense Breast Cancer Idea Award [BC102152]; a National Science Foundation CAREER award to Lammerding J [CBET-1254846]; and a Pilot Project Award by the Cornell Center on the Microenvironment & Metastasis through Award Number U54CA143876 from the National Cancer Institute. The content of this article is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute or the National Institutes of Health.

Conflicts of Interest

Patricia M. Davidson, Celine Denais, Maya C. Bakshi, and Jan Lammerding declare that they have no conflicts of interest.

Ethical Standards

No human studies were carried out by the authors for this article. No animal studies were carried out by the authors for this article.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jan Lammerding.

Additional information

Associate Editor David Schaffer oversaw the review of this article.

This paper is part of the 2014 Young Innovators Issue.

Dr. Jan Lammerding is an Assistant Professor in the Department of Biomedical Engineering and the Weill Institute for Cell and Molecular Biology at Cornell University. He received a Bachelor of Engineering degree from the Thayer School of Engineering at Dartmouth College, a Diplom Ingenieur degree in Mechanical Engineering from the University of Technology Aachen, Germany, and a Ph.D. in Biological Engineering from the Massachusetts Institute of Technology (MIT). Before joining Cornell University, Dr. Lammerding served as a faculty member at Harvard Medical School/Brigham and Women’s Hospital (BWH) while also teaching in the Department of Biological Engineering at MIT. At Cornell, the Lammerding laboratory is developing novel experimental techniques to investigate the interplay between cellular mechanics and function, with a particular emphasis on the cell nucleus and its response to mechanical forces. Dr. Lammerding has won several prestigious awards, including a National Science Foundation CAREER Award, an American Heart Association Scientist Development Grant, and the BWH Department of Medicine Young Investigator Award. Dr. Lammerding has published over 40 peer-reviewed articles, including in Nature and PNAS. His research is supported by grants from the National Institutes of Health, the National Science Foundation, the Department of Defense Breast Cancer Research Program, the American Heart Association, and the Progeria Research Foundation.

figure a

Electronic supplementary material

Below is the link to the electronic supplementary material.

12195_2014_342_MOESM1_ESM.avi

Supplemental Video 1: Lmna +/+ cell migrating through a narrow constriction. Representative time-lapse video of a wild-type (Lmna +/+) mouse embryonic fibroblast migrating through a 3 × 5 µm2 constriction. To aid the viewer, the nucleus is outlined with a dashed white line at the beginning of the video. The vertical white lines indicate the beginning (left) and end (right) of the constriction, as defined for the migration transit time measurements (compare with Figure 4). Time measurements (indicated in the top left corner of the image) are based relative to the nucleus entering the constriction. In this video, the migration transit time was 3:20 h. The time interval between images was 10 minutes; scale bar: 25 µm. (AVI 629 kb)

12195_2014_342_MOESM2_ESM.avi

Supplemental Video 2: Lmna +/– cell migrating through a narrow constriction. Representative time-lapse video of a Lmna +/– mouse embryonic fibroblast migrating through a 3 × 5 µm2 constriction. To aid the viewer, the nucleus is outlined with a dashed white line at the beginning of the video. The vertical white lines indicate the beginning (left) and end (right) of the constriction, as defined for the migration transit time measurements (compare with Figure 4). Time measurements (indicated in the top left corner of the image) are based relative to the nucleus entering the constriction. In this video, the migration transit time was 1:40 h. The time interval between images was 10 minutes; scale bar: 25 µm. (AVI 594 kb)

12195_2014_342_MOESM3_ESM.avi

Supplemental Video 3: Lmna –/– cell migrating through a narrow constriction. Representative time-lapse video of a lamin A/C-deficient (Lmna –/–) mouse embryonic fibroblast migrating through a 3 × 5 µm2 constriction. To aid the viewer, the nucleus is outlined with a dashed white line at the beginning of the video. The vertical white lines indicate the beginning (left) and end (right) of the constriction, as defined for the migration transit time measurements (compare with Figure 4). Time measurements (indicated in the top left corner of the image) are based relative to the nucleus entering the constriction. In this video, the migration transit time was 1:40 h. The time interval between images was 10 minutes; scale bar: 25 µm. (AVI 675 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Davidson, P.M., Denais, C., Bakshi, M.C. et al. Nuclear Deformability Constitutes a Rate-Limiting Step During Cell Migration in 3-D Environments. Cel. Mol. Bioeng. 7, 293–306 (2014). https://doi.org/10.1007/s12195-014-0342-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12195-014-0342-y

Keywords

Navigation