Review
Cellular mechanosensing: Getting to the nucleus of it all

https://doi.org/10.1016/j.pbiomolbio.2014.06.009Get rights and content

Abstract

Cells respond to mechanical forces by activating specific genes and signaling pathways that allow the cells to adapt to their physical environment. Examples include muscle growth in response to exercise, bone remodeling based on their mechanical load, or endothelial cells aligning under fluid shear stress. While the involved downstream signaling pathways and mechanoresponsive genes are generally well characterized, many of the molecular mechanisms of the initiating ‘mechanosensing’ remain still elusive. In this review, we discuss recent findings and accumulating evidence suggesting that the cell nucleus plays a crucial role in cellular mechanotransduction, including processing incoming mechanoresponsive signals and even directly responding to mechanical forces. Consequently, mutations in the involved proteins or changes in nuclear envelope composition can directly impact mechanotransduction signaling and contribute to the development and progression of a variety of human diseases, including muscular dystrophy, cancer, and the focus of this review, dilated cardiomyopathy. Improved insights into the molecular mechanisms underlying nuclear mechanotransduction, brought in part by the emergence of new technologies to study intracellular mechanics at high spatial and temporal resolution, will not only result in a better understanding of cellular mechanosensing in normal cells but may also lead to the development of novel therapies in the many diseases linked to defects in nuclear envelope proteins.

Introduction

Mechanotransduction describes the cellular and molecular processes of converting mechanical stimuli into biochemical signals. Since the discovery of stretch sensitive ion channels, the field has rapidly expanded, leading to the discovery of various force sensitive proteins within the cytoplasm and plasma membrane, such as titin, talin, vinculin, and p130Cas (Seifert and Grater, 2013). Disturbed cellular mechanotransduction causes numerous defects on the cell, tissue, and organ level (Jaalouk and Lammerding, 2009); thus, it only seems logical that the human heart, which beats on average 2.5 billion times over the course of a lifetime, requires tightly regulated and robust mechanoregulation. Beyond this, the importance of mechanoelectric feedback adds another layer of complexity to cardiac mechanotransduction. For instance, ion channels such as Cav1.2 and the TRP family of ion channels have specific locations in the heart and enable organ level responses to pressure and volume fluctuations by helping regulate action potentials (Takahashi et al., 2013). Connexins, transmembrane proteins important for gap junction formation, may act as effectors to coordinate excitation-contraction coupling (Meens et al., 2013). Beyond this, connexin-43 is upregulated in response to mechanical stimulation and precedes other cell–cell junction formations. While mechanosensing at the plasma membrane and the cytoskeleton has been well studied, our knowledge quickly diminishes as we probe deeper into (cardiac) cells. It is clear that cardiac myocyte nuclei undergo substantial deformations during contraction (Fig. 1); however, one remaining central question in cardiac mechanobiology and mechanobiology in general, revolves around the extent to which the nucleus itself can act as a mechanosensor.

A mechanosensor, as we will define it, is a protein (or, more generally, a cellular structure) that translates a mechanical input into a biochemical output, thereby initiating mechanoresponsive signaling pathways. Typically, molecular mechanosensing involves force induced conformational changes, resulting in altered interaction with binding partners or modulation of (protein) activity. It is well established that the nucleus plays an important role in mechanotransduction signaling, i.e., the process of biochemical signal propagation and processing, as most signaling pathways eventually culminate with nuclear proteins binding to specific genomic elements to modulate transcription. It is also known that the nucleus is mechanically connected to the rest of the cell via LINC (Linker of Nucleoskeleton and Cytoskeleton) complex structures in the nuclear envelope. These complexes are akin to focal adhesions at the plasma membrane, allowing for cytoskeletal and external forces to result in nuclear deformations (Lombardi and Lammerding, 2011). But, the question remains: can mechanically induced nuclear deformations directly control gene expression in a predictable, biologically-meaningful way? If so, what are the molecular mechanisms that enable the nucleus to sense and respond in this way? What are the implications for cardiac function in health and disease? Are there cardiac specific mechanisms for nuclear mechanotransduction?

The emergence of new technologies–ranging from advanced imaging and bioengineering approaches for cell-based assays, to molecular probes that can detect nanoscale forces and deformations–has enabled the scientific community to finally start addressing this important question. In the following sections, we will briefly outline current models of nuclear mechanotransduction before discussing how recent findings, ranging from cellular and subcellular studies to human diseases, and corresponding disease models are beginning to shape a clearer portrait of the nucleus as both a mechanosensor and a central processing hub for mechanoresponsive signaling pathways. Lastly, we will highlight how recent and ongoing advances in technology development will help to further elucidate the fascinating biology of nuclear mechanotransduction and its role in human health and disease.

Section snippets

The nucleus at the center of the cell

A basic overview of the mechanical connectivity linking the nucleus to the rest of the cell and its ECM surroundings (Fig. 2) provides a helpful roadmap for understanding how the nucleus may carry out mechanotransduction processes. The nucleus is encapsulated by a double lipid membrane system, composed of the inner and outer nuclear membrane (INM, ONM), that is studded with nuclear pores. Driven by recent advances in protein isolation, mass spectroscopy, and super-resolution imaging, it is now

The theory – potential mechanisms of nuclear mechanosensing

As we gain a better understanding of the structure and function of the nuclear envelope and interior, we move ever closer to comprehending how the nucleus perceives and responds to force. Based on our current knowledge, a number of (non-mutually exclusive) molecular mechanisms have been proposed that could directly modulate nuclear structure and transcriptional regulation, thereby enabling the nucleus to transduce mechanical forces into biochemical signals (Fig. 3).

The case for nuclear mechanotransduction – getting to the nucleus of it

Cells subjected to mechanical stimuli respond with rapid (<30 min) activation of characteristic ‘mechanosensitive’ genes such as Egr-1, Iex-1, or Pai-1 that often represent immediate early transcription factors that turn on additional genes. Many of these responses appear quite ubiquitous and can be observed in a number of different cell lines, including fibroblasts, myotubes, and neonatal cardiac myocytes (Banerjee et al., 2014, Ho et al., 2013b, Lammerding et al., 2004). Intriguingly, cells

Mutations in nuclear envelope proteins cause dilated cardiomyopathy

Achieving an understanding of the mechanisms underlying human disease is one of the major drivers in biomedical sciences. A powerful illustration of the importance of mechanotransduction comes from the large number of human diseases linked to defects in cellular structure and mechanosensing, including cardiomyopathies, vascular disease, muscular dystrophy, cancer, reversal of the inner organs, and hearing defects (Jaalouk and Lammerding, 2009). Muscle diseases, such as cardiomyopathies and

The road ahead – technology leading the way

The ability to identify specific changes to gene location, chromatin structure, chromatin organization, and transcriptional activity in response to mechanical force would bring much clarity to the specific nuclear mechanotransduction mechanisms at work. Emerging methods and technologies that can relate intranuclear forces with biochemical events such as conformational changes and transcriptional processes, particularly on the single cell level, have the potential to transform the way we study

Outlook & conclusions

We can confidently proclaim that the nucleus plays an important role in cellular mechanotransduction and function, as the many diseases resulting from mutations in nuclear envelope proteins attest. Increasing evidence suggests that the nucleus itself can respond to mechanical forces and deformations, resulting in altered transcriptional regulation and cellular function. However, many of the mechanisms remain unclear and underexplored, including the precise role of nuclear envelope proteins such

Editors’ note

Please see also related communications in this issue by Vostarek et al. (2014) and Dhein et al. (2014).

Acknowledgments

The authors apologize to all authors whose work could not be cited due to space constraints. This work was supported by National Institutes of Health awards [R01 HL082792 and R01 NS59348]; a 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, as well as NSF

References (193)

  • J.I. de las Heras et al.

    Cancer biology and the nuclear envelope: a convoluted relationship

    Semin. Cancer Biol.

    (2013)
  • S. Deguchi et al.

    Flow-induced hardening of endothelial nucleus as an intracellular stress-bearing organelle

    J. Biomech.

    (2005)
  • J. Demmerle et al.

    The nuclear envelope protein emerin binds directly to histone deacetylase 3 (HDAC3) and activates HDAC3 activity

    J. Biol. Chem.

    (2012)
  • L.J. Emerson et al.

    Defects in cell spreading and ERK1/2 activation in fibroblasts with lamin A/C mutations

    Biochim. Biophys. Acta (BBA)-Mol. Basis Dis.

    (2009)
  • F. Fagotto et al.

    Nuclear localization signal-independent and importin/karyopherin-independent nuclear import of beta-catenin

    Curr. Biol.

    (1998)
  • J.D. Finan et al.

    Osmotic stress alters chromatin condensation and nucleocytoplasmic transport

    Biochem. Biophys. Res. Commun.

    (2011)
  • F. Guilak

    Compression-induced changes in the shape and volume of the chondrocyte nucleus

    J. Biomech.

    (1995)
  • F. Guilak et al.

    The mechanical environment of the chondrocyte: a biphasic finite element model of cell-matrix interactions in articular cartilage

    J. Biomech.

    (2000)
  • G.G. Gundersen et al.

    Nuclear positioning

    Cell

    (2013)
  • C.M. Hale et al.

    Dysfunctional connections between the nucleus and the actin and microtubule networks in laminopathic models

    Biophys. J.

    (2008)
  • L. Hernandez et al.

    Functional coupling between the extracellular matrix and nuclear lamina by wnt signaling in progeria

    Dev. Cell

    (2010)
  • S. Hihara et al.

    Local nucleosome dynamics facilitate chromatin accessibility in living mammalian cells

    Cell Rep.

    (2012)
  • J.M. Holaska et al.

    Transcriptional repressor germ cell-less (GCL) and barrier to autointegration factor (BAF) compete for binding to emerin in vitro

    J. Biol. Chem.

    (2003)
  • S. Hübner et al.

    Laminopathy-inducing lamin A mutants can induce redistribution of lamin binding proteins into nuclear aggregates

    Exp. Cell Res.

    (2006)
  • A. Inoue et al.

    Perturbation of HP1 localization and chromatin binding ability causes defects in sister-chromatid cohesion

    Mutat. Res. Gen. Toxicol. Environ. Mutagen.

    (2008)
  • P. Isermann et al.

    Nuclear mechanics and mechanotransduction in health and disease

    Curr. Biol.

    (2013)
  • K.V. Iyer et al.

    Mechanical activation of cells induces chromatin remodeling preceding MKL nuclear transport

    Biophys. J.

    (2012)
  • B. Kalverda et al.

    Nucleoporins directly stimulate expression of developmental and cell-cycle genes inside the nucleoplasm

    Cell

    (2010)
  • M. Knight et al.

    Cell and nucleus deformation in compressed chondrocyte–alginate constructs: temporal changes and calculation of cell modulus

    Biochim. Biophys. Acta (BBA)-Gen. Subj.

    (2002)
  • T. Kohl et al.

    Superresolution microscopy in heart – cardiac nanoscopy

    J. Mol. Cell. Cardiol.

    (2013)
  • J. Lammerding et al.

    Lamins A and C but not lamin B1 regulate nuclear mechanics

    J. Biol. Chem.

    (2006)
  • J.S.H. Lee et al.

    Nuclear lamin A/C deficiency induces defects in cell mechanics, polarization, and migration

    Biophys. J.

    (2007)
  • Q.S. Li et al.

    The regulation of dynamic mechanical coupling between actin cytoskeleton and nucleus by matrix geometry

    Biomaterials

    (2014)
  • Y. Li et al.

    Biophysical regulation of histone acetylation in mesenchymal stem cells

    Biophys. J.

    (2011)
  • F. Lin et al.

    MAN1, an inner nuclear membrane protein that shares the LEM domain with lamina-associated polypeptide 2 and emerin

    J. Biol. Chem.

    (2000)
  • B. Liu et al.

    Resveratrol rescues SIRT1-dependent adult stem cell decline and alleviates progeroid features in laminopathy-based progeria

    Cell metab.

    (2012)
  • M.L. Lombardi et al.

    The interaction between nesprins and sun proteins at the nuclear envelope is critical for force transmission between the nucleus and cytoskeleton

    J. Biol. Chem.

    (2011)
  • A. Akhtar et al.

    The nuclear envelope and transcriptional control

    Nat. Rev. Genet.

    (2007)
  • V. Andres et al.

    Role of A-type lamins in signaling, transcription, and chromatin organization

    J. Cell. Biol.

    (2009)
  • T. Arimura et al.

    Mouse model carrying H222P-Lmna mutation develops muscular dystrophy and dilated cardiomyopathy similar to human striated muscle laminopathies

    Hum. Mol. Genet.

    (2005)
  • F. Azibani et al.

    Striated muscle laminopathies

  • C. Baarlink et al.

    Nuclear actin network assembly by formins regulates the SRF coactivator MAL

    Science

    (2013)
  • E.M. Balzer et al.

    Physical confinement alters tumor cell adhesion and migration phenotypes

    FASEB J.

    (2012)
  • A. Banerjee et al.

    Viscoelastic behavior of human lamin a proteins in the context of dilated cardiomyopathy

    PLoS One

    (2013)
  • I. Banerjee et al.

    Targeted ablation of nesprin 1 and nesprin 2 from murine myocardium results in cardiomyopathy, altered nuclear morphology and inhibition of the biomechanical gene response

    PLoS Genet.

    (2014)
  • D.Z. Bar et al.

    BAF-1 mobility is regulated by environmental stresses

    Mol. biol. Cell.

    (2014)
  • B.J. Belin et al.

    Visualization of actin filaments and monomers in somatic cell nuclei

    Mol. Biol. Cell.

    (2013)
  • L. Bengtsson et al.

    LUMA interacts with emerin and influences its distribution at the inner nuclear membrane

    J. Cell Sci.

    (2008)
  • J.J. Benitez et al.

    Microfluidic extraction, stretching and analysis of human chromosomal DNA from single cells

    Lab. Chip

    (2012)
  • S. Bione et al.

    Identification of a novel X-linked gene responsible for Emery-Dreifuss muscular-dystrophy

    Nat. Genet.

    (1994)
  • Cited by (139)

    • Cancer cell mechanobiology: a new frontier for cancer research

      2022, Journal of the National Cancer Center
      Citation Excerpt :

      Often, the ability to invade is the threshold that separates precancerous lesions from cancerous lesions. The underlying molecular mechanisms for regulating cellular mechanotypes are rather elusive, though it is accepted that complex biological processes, such as pathways associated with cell adhesion, cytoskeletal remodeling, epithelial-mesenchymal transition (EMT), and the nucleoskeleton, are involved 1–3. In recent years, emerging evidence has shown that cell mechanotypical changes, such as alterations in cellular stiffness and deformability, are common phenotypic events in cancer development and progression.

    View all citing articles on Scopus
    View full text