Cell volume change through water efflux impacts cell stiffness and stem cell fate

Significance Cell volume is thought to be a well-controlled cellular characteristic, increasing as a cell grows, while macromolecular density is maintained. We report that cell volume can also change in response to external physical cues, leading to water influx/efflux, which causes significant changes in subcellular macromolecular density. This is observed when cells spread out on a substrate: Cells reduce their volume and increase their molecular crowding due to an accompanying water efflux. Exploring this phenomenon further, we removed water from mesenchymal stem cells through osmotic pressure and found this was sufficient to alter their differentiation pathway. Based on these results, we suggest cells chart different differentiation and behavioral pathways by sensing/altering their cytoplasmic volume and density through changes in water influx/efflux. Cells alter their mechanical properties in response to their local microenvironment; this plays a role in determining cell function and can even influence stem cell fate. Here, we identify a robust and unified relationship between cell stiffness and cell volume. As a cell spreads on a substrate, its volume decreases, while its stiffness concomitantly increases. We find that both cortical and cytoplasmic cell stiffness scale with volume for numerous perturbations, including varying substrate stiffness, cell spread area, and external osmotic pressure. The reduction of cell volume is a result of water efflux, which leads to a corresponding increase in intracellular molecular crowding. Furthermore, we find that changes in cell volume, and hence stiffness, alter stem-cell differentiation, regardless of the method by which these are induced. These observations reveal a surprising, previously unidentified relationship between cell stiffness and cell volume that strongly influences cell biology.

[1]  Gwo‐Jaw Wang,et al.  Steroid effects on osteogenesis through mesenchymal cell gene expression , 2004, Osteoporosis International.

[2]  Megan N. McClean,et al.  Severe osmotic compression triggers a slowdown of intracellular signaling, which can be explained by molecular crowding , 2013, Proceedings of the National Academy of Sciences.

[3]  Ian A. Swinburne,et al.  Interplay of Cell Shape and Division Orientation Promotes Robust Morphogenesis of Developing Epithelia , 2014, Cell.

[4]  Miroslav Zivkovic,et al.  A finite element model of cell deformation during magnetic bead twisting. , 2002, Journal of applied physiology.

[5]  Sirio Dupont Role of YAP/TAZ in mechanotransduction , 2011 .

[6]  D. Weitz,et al.  Elastic Behavior of Cross-Linked and Bundled Actin Networks , 2004, Science.

[7]  T. Pederson,et al.  COMPARISON OF MITOTIC PHENOMENA AND EFFECTS INDUCED BY HYPERTONIC SOLUTIONS IN HELA CELLS , 1970, The Journal of cell biology.

[8]  P. Janmey,et al.  Tissue Cells Feel and Respond to the Stiffness of Their Substrate , 2005, Science.

[9]  M. Bartoo,et al.  The stiffness of rabbit skeletal actomyosin cross-bridges determined with an optical tweezers transducer. , 1998, Biophysical journal.

[10]  P. Arratia,et al.  Absence of filamin A prevents cells from responding to stiffness gradients on gels coated with collagen but not fibronectin. , 2009, Biophysical journal.

[11]  Dirar Homouz,et al.  Structure, function, and folding of phosphoglycerate kinase are strongly perturbed by macromolecular crowding , 2010, Proceedings of the National Academy of Sciences.

[12]  Y. Wang,et al.  Cell locomotion and focal adhesions are regulated by substrate flexibility. , 1997, Proceedings of the National Academy of Sciences of the United States of America.

[13]  D. Häussinger,et al.  Functional significance of cell volume regulatory mechanisms. , 1998, Physiological reviews.

[14]  Y. Wang,et al.  High resolution detection of mechanical forces exerted by locomoting fibroblasts on the substrate. , 1999, Molecular biology of the cell.

[15]  D. Weitz,et al.  Micron-scale coherence in interphase chromatin dynamics , 2013, Proceedings of the National Academy of Sciences.

[16]  Y. Jan,et al.  Ankyrin Repeats Convey Force to Gate the NOMPC Mechanotransduction Channel , 2015, Cell.

[17]  R. Ellis Macromolecular crowding : obvious but underappreciated , 2022 .

[18]  G. Whitesides,et al.  Soft Lithography. , 1998, Angewandte Chemie.

[19]  Harald Sontheimer,et al.  Chloride accumulation drives volume dynamics underlying cell proliferation and migration. , 2009, Journal of neurophysiology.

[20]  Fei Wang,et al.  Material Properties of the Cell Dictate Stress-induced Spreading and Differentiation in Embryonic Stem Cells Growing Evidence Suggests That Physical Microenvironments and Mechanical Stresses, in Addition to Soluble Factors, Help Direct Mesenchymal-stem-cell Fate. However, Biological Responses to a L , 2022 .

[21]  D. Navajas,et al.  Scaling the microrheology of living cells. , 2001, Physical review letters.

[22]  Yangqing Xu,et al.  Development of time-integrated multipoint moment analysis for spatially resolved fluctuation spectroscopy with high time resolution. , 2011, Biophysical Journal.

[23]  Yongjin Sung,et al.  Multiple Phases of Chondrocyte Enlargement Underlie Differences in Skeletal Proportions , 2013, Nature.

[24]  Donald E. Ingber,et al.  Activation of Mechanosensitive Ion Channels by Forces Transmitted Through Integrins and the Cytoskeleton , 2007 .

[25]  G. Charras,et al.  The cytoplasm of living cells behaves as a poroelastic material , 2013, Nature materials.

[26]  W. Möller,et al.  Improved spinning top aerosol-generator for the production of high concentrated ferrimagnetic aerosols , 1990 .

[27]  Ning Wang,et al.  Spatial and temporal traction response in human airway smooth muscle cells. , 2002, American journal of physiology. Cell physiology.

[28]  Philip Ball,et al.  Water as an Active Constituent in Cell Biology , 2008 .

[29]  Harald Sontheimer,et al.  Hydrodynamic Cellular Volume Changes Enable Glioma Cell Invasion , 2011, The Journal of Neuroscience.

[30]  S. Sen,et al.  Matrix Elasticity Directs Stem Cell Lineage Specification , 2006, Cell.

[31]  Anthony A. Hyman,et al.  Quantification of surface tension and internal pressure generated by single mitotic cells , 2014, Scientific Reports.

[32]  Hiroshi Kimura,et al.  Kinetics of Core Histones in Living Human Cells , 2001, The Journal of cell biology.

[33]  Ming Guo,et al.  Probing the Stochastic, Motor-Driven Properties of the Cytoplasm Using Force Spectrum Microscopy , 2014, Cell.

[34]  C F Dewey,et al.  Theoretical estimates of mechanical properties of the endothelial cell cytoskeleton. , 1996, Biophysical journal.

[35]  Kheya Sengupta,et al.  Fibroblast adaptation and stiffness matching to soft elastic substrates. , 2007, Biophysical journal.

[36]  Donald E Ingber,et al.  Micropatterning tractional forces in living cells. , 2002, Cell motility and the cytoskeleton.

[37]  Dennis E. Discher,et al.  Nuclear Lamin-A Scales with Tissue Stiffness and Enhances Matrix-Directed Differentiation , 2013, Science.

[38]  C. R. Ethier,et al.  Altered mechanobiology of Schlemm’s canal endothelial cells in glaucoma , 2014, Proceedings of the National Academy of Sciences.

[39]  P. Janmey,et al.  Elasticity of semiflexible biopolymer networks. , 1995, Physical review letters.

[40]  Rob Phillips,et al.  Membrane-protein interactions in mechanosensitive channels. , 2004, Biophysical journal.

[41]  A. Minton,et al.  The Influence of Macromolecular Crowding and Macromolecular Confinement on Biochemical Reactions in Physiological Media* , 2001, The Journal of Biological Chemistry.

[42]  Adhesive micropatterns for cells: a microcontact printing protocol. , 2009, Cold Spring Harbor protocols.

[43]  C. Broedersz,et al.  Origins of elasticity in intermediate filament networks. , 2010, Physical review letters.

[44]  M. Kirschner,et al.  Cell Growth and Size Homeostasis in Proliferating Animal Cells , 2009, Science.

[45]  Bert Poolman,et al.  A sensor for quantification of macromolecular crowding in living cells , 2015, Nature Methods.

[46]  C. S. Chen,et al.  Geometric control of cell life and death. , 1997, Science.

[47]  Timothy J Mitchison,et al.  Dissecting Temporal and Spatial Control of Cytokinesis with a Myosin II Inhibitor , 2003, Science.

[48]  Christopher S. Chen,et al.  Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. , 2004, Developmental cell.

[49]  P. Janmey,et al.  Actin-binding protein requirement for cortical stability and efficient locomotion. , 1992, Science.

[50]  D A Weitz,et al.  Universal behavior of the osmotically compressed cell and its analogy to the colloidal glass transition , 2009, Proceedings of the National Academy of Sciences.

[51]  David J. Mooney,et al.  Harnessing Traction-Mediated Manipulation of the Cell-Matrix Interface to Control Stem Cell Fate , 2010, Nature materials.

[52]  Dennis Discher,et al.  Substrate compliance versus ligand density in cell on gel responses. , 2004, Biophysical journal.

[53]  E. Hoffmann,et al.  Physiology of cell volume regulation in vertebrates. , 2009, Physiological reviews.

[54]  D A Weitz,et al.  Filamin A is essential for active cell stiffening but not passive stiffening under external force. , 2009, Biophysical journal.

[55]  M. Knight,et al.  Osmotic challenge drives rapid and reversible chromatin condensation in chondrocytes. , 2013, Biophysical journal.

[56]  J J Fredberg,et al.  Selected contribution: time course and heterogeneity of contractile responses in cultured human airway smooth muscle cells. , 2001, Journal of applied physiology.

[57]  J. Swanson,et al.  Cellular dimensions affecting the nucleocytoplasmic volume ratio , 1991, The Journal of cell biology.

[58]  David A Weitz,et al.  The role of vimentin intermediate filaments in cortical and cytoplasmic mechanics. , 2013, Biophysical journal.

[59]  Ben Fabry,et al.  Traction fields, moments, and strain energy that cells exert on their surroundings. , 2002, American journal of physiology. Cell physiology.

[60]  Daniel J. Muller,et al.  Hydrostatic pressure and the actomyosin cortex drive mitotic cell rounding , 2011, Nature.