Trends in Neurosciences
Volume 44, Issue 2, February 2021, Pages 110-121
Journal home page for Trends in Neurosciences

Review
Mechanical Forces Orchestrate Brain Development

https://doi.org/10.1016/j.tins.2020.10.012Get rights and content

Highlights

  • Mechanotransduction of environmental signals governs important cell functions during brain development.

  • Recent discoveries have shown that disruption of brain mechanical properties during embryogenesis is associated with neurodevelopmental disorders. Thus, a better understanding of mechanobiology will also enhance our knowledge of brain disorders.

  • The advent of new technologies will increase our ability to measure and manipulate the physical features of neuronal cells and their environment.

During brain development, progenitors generate successive waves of neurons that populate distinct cerebral regions, where they settle and differentiate within layers or nuclei. While migrating and differentiating, neurons are subjected to mechanical forces arising from the extracellular matrix, and their interaction with neighboring cells. Changes in brain biomechanical properties, during its formation or aging, are converted in neural cells by mechanotransduction into intracellular signals that control key neurobiological processes. Here, we summarize recent findings that support the contribution of mechanobiology to neurodevelopment, with focus on the cerebral cortex. Also discussed are the existing toolbox and emerging technologies made available to assess and manipulate the physical properties of neurons and their environment.

Section snippets

Mechanotransduction and Brain Development

The morphogenic changes that occur during brain development involve cell movement and cell shape remodeling that exert mechanical forces on neighboring cells and their extracellular matrix (ECM). The maturation and differentiation of neurons partly relies on transduction of those mechanical forces into intracellular biochemical signals via a process called mechanotransduction (see Glossary). Several molecular actors have been involved in this process and include for instance, adhesion molecules

Changes in Brain Biomechanical Properties Drive Developmental Processes

The brain is one of the softest tissues of the body, whose mechanical properties rely on both the molecular composition of its ECM (e.g., weak expression of collagen I) and intracellular components (e.g., low content of nuclear lamin A) [5]. The brain ECM proteome (matrisome) is predominantly composed of hyaluronic acid and proteoglycans [6], and its qualitative changes contribute to spatio-temporal modifications of ECM elasticity during development. As a result of the stabilization of their

Mechanotransduction as an Additional Layer of Neuronal Regulation

In general, cells convert biomechanical stimuli into intracellular signals via mechanotransduction. This process is shared by adult neurons, such as the peripheral ones of the somatosensory system that are sensitive to mechanical stretching, compression, vibration, or touch [24], and immature neurons, whose growth cones read mechanical cues [25]. However, in most cases, the underlying mechanisms by which neurons integrate mechanical information remain elusive.

Biomechanical Control of Early Neuronal Functions in Health and Disease

Despite its relative softness, the cerebral cortical wall shows a positive apico-basal stiffness gradient along its differentiation axis [8], which suggests a possible contribution of surrounding mechanical forces to neurogenesis. Indeed, myosin II generates local membrane tensions in response to traction forces causing matrix deformation that, by activating Piezo1 in cultured human neural stem cells (hNSCs), favors the generation of neurons [38]. Additional evidence obtained in rodents, shows

Concluding Remarks

The mechanical properties of the cerebral cortex vary between different locations and over time, and neural cells sense and respond dynamically to these changes. Activation of specific mechanotransduction pathways, instructs local morphogenetic events in neurons ranging from filopodia formation to synaptic maturation. Accumulating data indicate that cell migration contributes to brain morphogenesis. The softness traits of neurons make them susceptible to nuclear deformation upon mechanical

Acknowledgments

The work in the Nguyen laboratory is supported by the F.R.S.-F.N.R.S. (Synet; EOS 0019118F-RG36), the Fonds Leon Fredericq, the Fondation Médicale Reine Elisabeth, the Fondation Simone et Pierre Clerdent, the Belgian Science Policy (IAP-VII network P7/20), and the ERANET Neuron STEM-MCD and NeuroTalk. M.J.-T. and L.N. are respectively postdoctoral researcher, and senior research associates of the F.R.S- F.N.R.S. All figures were created with BioRender.com.

Glossary

Bin/amphiphysin/Rvs (BAR)
family of proteins that contain a Bin/amphiphysin/Rvs (BAR) domain, which includes six subfamilies: N-BAR, BAR, F-BAR, I-BAR, PX-BAR, and BAR-PH. BAR proteins regulate the curvature of the cell membrane, and according to their structure they generate different membrane phenotypes.
Barrier-to-autointegration factor (BAF)
mammalian ATP-dependent chromatin remodeling complex that contains DNA and histone-binding domains.
Emerin
LEM domain-containing protein mainly localized in

References (99)

  • Z. Jahed

    Kindlin is mechanosensitive: force-induced conformational switch mediates cross-talk among integrins

    Biophys. J.

    (2019)
  • T. Takizawa

    The meaning of gene positioning

    Cell

    (2008)
  • H. Jing

    Exchange of GATA factors mediates transitions in looped chromatin organization at a developmentally regulated gene locus

    Mol. Cell

    (2008)
  • R.H. Nichol

    Environmental elasticity regulates cell-type specific RHOA signaling and neuritogenesis of human neurons

    Stem Cell Reports

    (2019)
  • I. Sack

    The impact of aging and gender on brain viscoelasticity

    NeuroImage

    (2009)
  • S. Varney

    Mice lacking integrin β3 expression exhibit altered response to chronic stress

    Neurobiol. Stress

    (2015)
  • C.A. Chaze

    Altered brain tissue viscoelasticity in pediatric cerebral palsy measured by magnetic resonance elastography

    Neuroimage Clin.

    (2019)
  • C. Gayrard et al.

    FRET-based molecular tension microscopy

    Methods

    (2016)
  • T. Mitchison et al.

    Cytoskeletal dynamics and nerve growth

    Neuron

    (1988)
  • K.M. Stroka

    Water permeation drives tumor cell migration in confined microenvironments

    Cell

    (2014)
  • J.M. Kalappurakkal

    Integrin mechano-chemical signaling generates plasma membrane nanodomains that promote cell spreading

    Cell

    (2019)
  • B. Yang

    Mechanosensing controlled directly by tyrosine kinases

    Nano Lett.

    (2016)
  • D.E. Leckband et al.

    Cadherin adhesion and mechanotransduction

    Annu. Rev. Cell Dev. Biol.

    (2014)
  • A.D. Stephens

    Chromatin and lamin A determine two different mechanical response regimes of the cell nucleus

    MBoC

    (2017)
  • J. Swift

    Nuclear Lamin-A scales with tissue stiffness and enhances matrix-directed differentiation

    Science

    (2013)
  • K.R. Long et al.

    How the extracellular matrix shapes neural development

    Open Biol.

    (2019)
  • M. Iwashita

    Systematic profiling of spatiotemporal tissue and cellular stiffness in the developing brain

    Development

    (2014)
  • A. Tanaka

    Regulation of neuritogenesis in hippocampal neurons using stiffness of extracellular microenvironment

    PLoS ONE

    (2018)
  • J. Kjell

    Defining the adult neural stem cell niche proteome identifies key regulators of adult neurogenesis

    Cell Stem Cell

    (2020)
  • C.D. Kroenke et al.

    How forces fold the cerebral cortex

    J. Neurosci.

    (2018)
  • C. Llinares-Benadero et al.

    Deconstructing cortical folding: genetic, cellular, and mechanical determinants

    Nat. Rev. Neurosci.

    (2019)
  • K.E. Garcia

    Dynamic patterns of cortical expansion during folding of the preterm human brain

    Proc. Natl. Acad. Sci. U. S. A.

    (2018)
  • S.A. Fietz

    Transcriptomes of germinal zones of human and mouse fetal neocortex suggest a role of extracellular matrix in progenitor self-renewal

    Proc. Natl. Acad. Sci. U. S. A.

    (2012)
  • E. Karzbrun

    Human brain organoids on a chip reveal the physics of folding

    Nat. Phys.

    (2018)
  • K.R. Long

    Extracellular matrix components HAPLN1, lumican, and collagen I, cause hyaluronic acid-dependent folding of the developing human neocortex

    Neuron

    (2018)
  • C. de Juan Romero

    Discrete domains of gene expression in germinal layers distinguish the development of gyrencephaly

    EMBO J.

    (2015)
  • D. del Toro

    Regulation of cerebral cortex folding by controlling neuronal migration via FLRT adhesion molecules

    Cell

    (2017)
  • P. Delmas

    Molecular mechanisms of mechanotransduction in mammalian sensory neurons

    Nat. Rev. Neurosci.

    (2011)
  • P.C. Kerstein

    Mechanochemical regulation of growth cone motility

    Front. Cell. Neurosci.

    (2015)
  • Z. Sun

    Integrin activation by talin, kindlin, and mechanical forces

    Nat. Cell Biol.

    (2019)
  • E. Klotzsch

    Fibronectin forms the most extensible biological fibers displaying switchable force-exposed cryptic binding sites

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • A. Elosegui-Artola

    Mechanical regulation of a molecular clutch defines force transmission and transduction in response to matrix rigidity

    Nat. Cell Biol.

    (2016)
  • T.-Y. Chang

    Paxillin facilitates timely neurite initiation on soft-substrate environments by interacting with the endocytic machinery

    eLife

    (2017)
  • E. Maffioli

    Proteomic dissection of nanotopography-sensitive mechanotransductive signaling hubs that foster neuronal differentiation in PC12 Cells

    Front. Cell. Neurosci.

    (2018)
  • S.W. Moore

    Netrin-1 attracts axons through FAK-dependent mechanotransduction

    J. Neurosci.

    (2012)
  • A. Reversat

    Cellular locomotion using environmental topography

    Nature

    (2020)
  • N. Srivastava

    Pressure sensing through Piezo channels controls whether cells migrate with blebs or pseudopods

    Proc. Natl. Acad. Sci. U. S. A.

    (2020)
  • S.-H. Woo

    Piezo2 is required for Merkel-cell mechanotransduction

    Nature

    (2014)
  • L. He

    Mechanical regulation of stem-cell differentiation by the stretch-activated Piezo channel

    Nature

    (2018)
  • Cited by (24)

    • Patterning of brain organoids derived from human pluripotent stem cells

      2022, Current Opinion in Neurobiology
      Citation Excerpt :

      To address these shortcomings, engineering solutions leveraging recent work in the fields of biomaterials, microfluidics and synthetic biology are increasingly being utilized [58–60]. Bioengineering approaches are uniquely suited for mimicking the complex array of biophysical cues [61,62] and morphogens [63,64] involved in regulating the developing CNS. A recent study used a combination of glass micropatterning of hiPSC cultures to spatially confine their growth with a 4% Matrigel environment to enable the formation of 3D cellular structures (Figure 3b).

    View all citing articles on Scopus
    View full text