Nuclear mechanics during cell migration

https://doi.org/10.1016/j.ceb.2010.10.015Get rights and content

During cell migration, the movement of the nucleus must be coordinated with the cytoskeletal dynamics at the leading edge and trailing end, and, as a result, undergoes complex changes in position and shape, which in turn affects cell polarity, shape, and migration efficiency. We here describe the steps of nuclear positioning and deformation during cell polarization and migration, focusing on migration through three-dimensional matrices. We discuss molecular components that govern nuclear shape and stiffness, and review how nuclear dynamics are connected to and controlled by the actin, tubulin and intermediate cytoskeleton-based migration machinery and how this regulation is altered in pathological conditions. Understanding the regulation of nuclear biomechanics has important implications for cell migration during tissue regeneration, immune defence and cancer.

Introduction

Cell migration is a complex physicochemical process which leads to translocation of the cell body across two-dimensional (2D) surfaces, through basement membranes, or through three-dimensional (3D) interstitial tissues [1]. During migration through 3D tissues, the stiffness and density of the surrounding extracellular matrix (ECM) present an additional physical challenge to the moving cell body. Two principal mechanisms are known by which migrating cells can overcome these constraints: firstly, proteolytic ECM degradation leading to gap widening and cell-generated trail formation and secondly, elastic and plastic deformations of the cell body to fit through the available space [2]. If a cell is unable to ‘squeeze’ through a particularly narrow region, it employs a third mechanism to maintain migration, that is, retraction of already established protrusions and repolarization to explore the adjacent environment for an alternative route, thereby bypassing the obstacle [3]. Consequently, translocation through 3D tissues is dependent on the deformability of both, the environment and the cell body.

The cell body consists of the plasma membrane surrounding the gel-like cytoplasm including small organelles (dimensions below the 1–2 μm range) and cytoskeletal filaments. These structures are morphologically adaptable enough to vigorously change and adjust to virtually any shape via a combination of firstly, fast (passive) mechanical deformation and secondly, slower cytoskeletal remodelling [4, 5•, 6]. Ultimately, cytoplasmic protrusions pass through gaps in the submicron range; this adaptability can be experimentally exploited to isolate protrusive cell regions including pseudopodia, filopodia and invadopodia using filters with pores sized of 1 μm and below [7, 8]. In contrast, the nucleus, the largest cellular organelle, is approximately 5–10 times stiffer than the surrounding cytoskeleton as it is mechanically stabilized by a constitutive network of structural proteins (see below); therefore it commonly resists large changes in shape [5•, 9, 10, 11, 12]. Consequently, for migration through small pores or 3D scaffolds, the nucleus can become the rate-limiting organelle [7, 13•, 14].

Nuclear shapes and sizes can vary widely between different cell types and even within cell lines; nonetheless, most cells imaged in situ or cultured in 3D substrates have ovoid or spherical shaped nuclei with diameters of 5–15 μm [15]. In 2D culture, cells often spread out significantly, resulting in more disk-shaped nuclei 10–20 μm in diameter and only a few micrometers in height [16]. In a few often highly mobile cell types, however, including myeloid and cancer cells, the nuclei are bean-shaped, lobulated or segmented (‘polymorphic’) (Table 1) and thus may develop greater morphological flexibility [14, 17] (Figure 1).

In vivo, the dimensions of structural pores available for migrating cells in basement membranes and interstitial connective tissue vary considerably, ranging from large gaps in the range of 30 μm and larger in loose connective tissue and lymph nodes [18, 19•] down to submicron pores in basement membranes [20]. For cells passing through pores which are smaller than the cell diameter, the extent of required cell deformation depends on whether the cell degrades the ECM proteolytically in regions of local compression, or not [21]. Surface-localized proteolytic cleavage of ECM fibers enlarges the available space and leads to the de novo formation of small tracks; the diameter of these tracks approximates the cross section of the cell and thereby reduces required cell deformation [13•, 22]. In both proteolytic and non-proteolytic migration through 3D tissues, the shapes of both cytoplasm und nucleus thus adopt their morphology and thereby minimize resistance towards tissue structures [3]. We here aim to integrate nuclear dynamics into the multistep model of cell migration through interstitial tissue and discuss the implications of nuclear mechanics for physiological and neoplastic cell migration and invasion.

Section snippets

Steps of cell migration

Dependent on whether proteases are utilized or not, cell migration in 3D environments consists of four or five respective steps which are executed in a concurrent and cyclic manner [1, 23] (Figure 2). First the cell polarizes by actin assembly into filaments which push the plasma membrane outward and form protrusions (step 1), followed by the interaction of cell protrusions with the extracellular tissue matrix (step 2). In proteolytic migration through 3D tissues, the proteolytic degradation

Molecular regulation of nuclear shape and rigidity

In interphase nuclei, the size, shape, and stiffness are determined by the nuclear envelope and the nuclear interior. The nuclear envelope consists of the inner and outer nuclear membrane, interconnected at the sites of nuclear pores, and the underlying nuclear lamina, a dense protein network mostly made of lamin proteins that are part of the intermediate filament family (Figure 3) [5].

Implications for cell dynamics in immune cell function and cancer

Alterations of the expression of proteins involved in nuclear morphology and plasticity occur during physiological cell differentiation and activation, as well as in deregulated form in cancer.

Conclusions

Through its connection to the actin, tubulin and intermediate filament cytoskeleton, the nucleus is a major mechanosensory integrator. Because of its size and high rigidity, compared to all other cell compartments, it further imposes a major physical challenge for cells moving in 3D environments. Consequently, studies on nuclear mechanics and its implications for cell migration should preferentially focus on 3D in vitro and in vivo models. Whereas increased nuclear deformability offers an

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

We gratefully acknowledge Uta Flucke for human soft tissue sarcoma and Willeke Blokx for melanoma samples, and Monika Zwerger for generating the lamin-modified MCF10A cells. This work was supported by the European Community (T3Net, European Training Network) to P.F., grant 917.10.364 by the Dutch Science Foundation (NWO) to K.W. and the National Institutes of Health grants HL082792 and NS059348 and the Cardiovascular Leadership Group Award to J.L.

References (96)

  • J. Schape et al.

    Influence of lamin A on the mechanical properties of amphibian oocyte nuclei measured by atomic force microscopy

    Biophys J

    (2009)
  • T. Shimi et al.

    The A- and B-type nuclear lamin networks: microdomains involved in chromatin organization and transcription

    Genes Dev

    (2008)
  • J. Lammerding et al.

    Lamin A/C deficiency causes defective nuclear mechanics and mechanotransduction

    J Clin Invest

    (2004)
  • M.S. Zastrow et al.

    Nuclear titin interacts with A- and B-type lamins in vitro and in vivo

    J Cell Sci

    (2006)
  • A.L. Olins et al.

    The human granulocyte nucleus: unusual nuclear envelope and heterochromatin composition

    Eur J Cell Biol

    (2008)
  • J.D. Pajerowski et al.

    Physical plasticity of the nucleus in stem cell differentiation

    Proc Natl Acad Sci U S A

    (2007)
  • Y.Y. Zhen et al.

    NUANCE a giant protein connecting the nucleus and actin cytoskeleton

    J Cell Sci

    (2002)
  • G. Wiche

    Role of plectin in cytoskeleton organization and dynamics

    J Cell Sci

    (1998)
  • K.J. Roux et al.

    Nesprin 4 is an outer nuclear membrane protein that can induce kinesin-mediated cell polarization

    Proc Natl Acad Sci U S A

    (2009)
  • F. Houben et al.

    Disturbed nuclear orientation and cellular migration in A-type lamin deficient cells

    Biochim Biophys Acta

    (2009)
  • M.N. Guilly et al.

    Lamins A/C are not expressed at early stages of human lymphocyte differentiation

    Exp Cell Res

    (1990)
  • J.S. Hale et al.

    Cell-extrinsic defective lymphocyte development in Lmna(−/−) mice

    PLoS One

    (2010)
  • P. Friedl et al.

    T cell migration in three-dimensional extracellular matrix: guidance by polarity and sensations

    Dev Immunol

    (2000)
  • M.E. Hudson et al.

    Identification of differentially expressed proteins in ovarian cancer using high-density protein microarrays

    Proc Natl Acad Sci U S A

    (2007)
  • N.D. Willis et al.

    Lamin A/C is a risk biomarker in colorectal cancer

    PLoS One

    (2008)
  • R.S. Venables et al.

    Expression of individual lamins in basal cell carcinomas of the skin

    Br J Cancer

    (2001)
  • A.J. Ridley et al.

    Cell migration: integrating signals from front to back

    Science

    (2003)
  • G. Bao et al.

    Cell and molecular mechanics of biological materials

    Nat Mater

    (2003)
  • K.N. Dahl et al.

    The nuclear envelope lamina network has elasticity and a compressibility limit suggestive of a molecular shock absorber

    J Cell Sci

    (2004)
  • O. Chaudhuri et al.

    Reversible stress softening of actin networks

    Nature

    (2007)
  • Z. Jia et al.

    Tumor cell pseudopodial protrusions. Localized signaling domains coordinating cytoskeleton remodeling, cell adhesion, glycolysis, RNA translocation, and protein translation

    J Biol Chem

    (2005)
  • N. Caille et al.

    Assessment of strain field in endothelial cells subjected to uniaxial deformation of their substrate

    Ann Biomed Eng

    (1998)
  • N. Caille et al.

    Contribution of the nucleus to the mechanical properties of endothelial cells

    J Biomech

    (2002)
  • C. Beadle et al.

    The role of myosin II in glioma invasion of the brain

    Mol Biol Cell

    (2008)
  • D.E. Jaalouk et al.

    Mechanotransduction gone awry

    Nat Rev Mol Cell Biol

    (2009)
  • S.B. Khatau et al.

    A perinuclear actin cap regulates nuclear shape

    Proc Natl Acad Sci U S A

    (2009)
  • D. Feng et al.

    Neutrophils emigrate from venules by a transendothelial cell pathway in response to FMLP

    J Exp Med

    (1998)
  • M. Bajenoff et al.

    Highways, byways and breadcrumbs: directing lymphocyte traffic in the lymph node

    Trends Immunol

    (2007)
  • K. Wolf et al.

    Collagen-based cell migration models in vitro and in vivo

    Semin Cell Dev Biol

    (2009)
  • P. Friedl et al.

    Migration of highly aggressive MV3 melanoma cells in 3-dimensional collagen lattices results in local matrix reorganization and shedding of alpha2 and beta1 integrins and CD44

    Cancer Res

    (1997)
  • P. Friedl et al.

    Proteolytic interstitial cell migration: a five-step process

    Cancer Metastasis Rev

    (2009)
  • A.J. Maniotis et al.

    Demonstration of mechanical connections between integrins, cytoskeletal filaments, and nucleoplasm that stabilize nuclear structure

    Proc Natl Acad Sci U S A

    (1997)
  • L.H. Tsai et al.

    Nucleokinesis in neuronal migration

    Neuron

    (2005)
  • J.R. Levy et al.

    Dynein drives nuclear rotation during forward progression of motile fibroblasts

    J Cell Sci

    (2008)
  • E.R. Gomes et al.

    Nuclear movement regulated by Cdc42, MRCK, myosin, and actin flow establishes MTOC polarization in migrating cells

    Cell

    (2005)
  • I. Dupin et al.

    Classical cadherins control nucleus and centrosome position and cell polarity

    J Cell Biol

    (2009)
  • L.P. Cramer

    Forming the cell rear first: breaking cell symmetry to trigger directed cell migration

    Nat Cell Biol

    (2010)
  • B.T. Schaar et al.

    Cytoskeletal coordination during neuronal migration

    Proc Natl Acad Sci U S A

    (2005)
  • Cited by (382)

    View all citing articles on Scopus
    View full text