We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Role of mechanical factors in fate decisions of stem cells

    Dong Li

    Department of Cell & Developmental Biology & Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA

    ,
    Jiaxi Zhou

    Department of Cell & Developmental Biology & Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA

    State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300200, China

    ,
    Farhan Chowdhury

    Department of Mechanical Science & Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA

    ,
    Jianjun Cheng

    Department of Materials Science & Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA

    ,
    Ning Wang

    Department of Mechanical Science & Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA

    &
    Published Online:https://doi.org/10.2217/rme.11.2

    Stem cells derived from adult tissues or from the inner cell mass of blastocyst-stage embryos can self-renew in culture and have the remarkable potential to undergo lineage-specific differentiation. Extensive studies have been devoted to achieving a better understanding of the soluble factors and the mechanism(s) by which they regulate the fate decisions of these cells, but it is only recently that a critical role has been revealed for physical and mechanical factors in controlling self-renewal and lineage specification. This review summarizes selected aspects of current work on stem cell mechanics with an emphasis on the influence of matrix stiffness, surface topography, cell shape and mechanical forces on the fate determination of mesenchymal stem cells and embryonic stem cells.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    Bibliography

    • Evans MJ, Kaufman MH: Establishment in culture of pluripotential cells from mouse embryos. Nature292,154–156 (1981).
    • Thomson JA, Itskovitz-Eldor J, Shapiro SS et al.: Embryonic stem cell lines derived from human blastocysts. Science282,1145–1147 (1998).
    • Jiang Y, Jahagirdar BN, Reinhardt RL et al.: Pluripotency of mesenchymal stem cells derived from adult marrow. Nature418,41–49 (2002).
    • Pittenger MF, Mackay AM, Beck SC et al.: Multilineage potential of adult human mesenchymal stem cells. Science284,143–147 (1999).
    • Deng J, Petersen BE, Steindler DA, Jorgensen ML, Laywell ED: Mesenchymal stem cells spontaneously express neural proteins in culture and are neurogenic after transplantation. Stem Cells24,1054–1064 (2006).
    • Hofstetter CP, Schwarz EJ, Hess D et al.: Marrow stromal cells form guiding strands in the injured spinal cord and promote recovery. Proc. Natl Acad. Sci. USA99,2199–2204 (2002).
    • Kondo T, Johnson SA, Yoder MC, Romand R, Hashino E: Sonic hedgehog and retinoic acid synergistically promote sensory fate specification from bone marrow-derived pluripotent stem cells. Proc. Natl Acad. Sci. USA102,4789–4794 (2005).
    • Engler AJ, Sen S, Sweeney HL, Discher DE: Matrix elasticity directs stem cell lineage specification. Cell126,677–689 (2006).▪▪ Matrix stiffness directs mesenchymal stem cell (MSC) lineage specification.
    • O’Neill C, Jordan P, Ireland G: Evidence for two distinct mechanisms of anchorage stimulation in freshly explanted and 3T3 Swiss mouse fibroblasts. Cell44,489–496 (1986).
    • 10  Chen CS, Mrksich M, Huang S, Whitesides GM, Ingber DE: Geometric control of cell life and death. Science276,1425–1428 (1997).
    • 11  Engler AJ, Griffin MA, Sen S, Bonnemann CG, Sweeney HL, Discher DE: Myotubes differentiate optimally on substrates with tissue-like stiffness: pathological implications for soft or stiff microenvironments. J. Cell. Biol.166,877–887 (2004).
    • 12  McBeath R, Pirone DM, Nelson CM, Bhadriraju K, Chen CS: Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Dev. Cell6,483–495 (2004).▪▪ Demonstrates that cell shape and RhoA activity control MSC differentiation.
    • 13  Georges PC, Miller WJ, Meaney DF, Sawyer ES, Janmey PA: Matrices with compliance comparable to that of brain tissue select neuronal over glial growth in mixed cortical cultures. Biophys. J.90,3012–3018 (2006).
    • 14  Discher DE, Janmey P, Wang YL: Tissue cells feel and respond to the stiffness of their substrate. Science310,1139–1143 (2005).
    • 15  Wang N, Tytell JD, Ingber DE: Mechanotransduction at a distance: mechanically coupling the extracellular matrix with the nucleus. Nat. Rev. Mol. Cell. Biol.10,75–82 (2009).
    • 16  Yeung T, Georges PC, Flanagan LA et al.: Effects of substrate stiffness on cell morphology, cytoskeletal structure, and adhesion. Cell Motil. Cytoskeleton60,24–34 (2005).
    • 17  Pouille PA, Ahmadi P, Brunet AC, Farge E: Mechanical signals trigger Myosin II redistribution and mesoderm invagination in Drosophila embryos. Sci. Signal2,ra16 (2009).
    • 18  Martin AC, Kaschube M, Wieschaus EF: Pulsed contractions of an actin-myosin network drive apical constriction. Nature457,495–499 (2009).
    • 19  Krieg M, Arboleda-Estudillo Y, Puech PH et al.: Tensile forces govern germ-layer organization in zebrafish. Nat. Cell Biol.10,429–436 (2008).
    • 20  Rauzi M, Verant P, Lecuit T, Lenne PF: Nature and anisotropy of cortical forces orienting Drosophila tissue morphogenesis. Nat. Cell Biol.10,1401–1410 (2008).
    • 21  Desprat N, Supatto W, Pouille PA, Beaurepaire E, Farge E: Tissue deformation modulates twist expression to determine anterior midgut differentiation in Drosophila embryos. Dev. Cell15,470–477 (2008).
    • 22  Levenstein ME, Ludwig TE, Xu RH et al.: Basic fibroblast growth factor support of human embryonic stem cell self-renewal. Stem Cells24,568–574 (2006).
    • 23  Vallier L, Alexander M, Pedersen RA: Activin/Nodal and FGF pathways cooperate to maintain pluripotency of human embryonic stem cells. J. Cell Sci.118,4495–4509 (2005).
    • 24  Xu RH, Sampsell-Barron TL, Gu F et al.: NANOG is a direct target of TGF-β/activin-mediated SMAD signaling in human ESCs. Cell Stem Cell3,196–206 (2008).
    • 25  Chadwick K, Wang L, Li L et al.: Cytokines and BMP-4 promote hematopoietic differentiation of human embryonic stem cells. Blood102,906–915 (2003).
    • 26  Xu RH, Chen X, Li DS et al.: BMP4 initiates human embryonic stem cell differentiation to trophoblast. Nat. Biotechnol.20,1261–1264 (2002).
    • 27  Zhang P, Li J, Tan Z et al.: Short-term BMP-4 treatment initiates mesoderm induction in human embryonic stem cells. Blood111,1933–1941 (2008).
    • 28  D’Amour KA, Agulnick AD, Eliazer S, Kelly OG, Kroon E, Baetge EE: Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat. Biotechnol.23,1534–1541 (2005).
    • 29  Chambers SM, Fasano CA, Papapetrou EP, Tomishima M, Sadelain M, Studer L: Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat. Biotechnol.27,275–280 (2009).
    • 30  Pera MF, Andrade J, Houssami S et al.: Regulation of human embryonic stem cell differentiation by BMP-2 and its antagonist noggin. J. Cell Sci.117,1269–1280 (2004).
    • 31  Smith JR, Vallier L, Lupo G, Alexander M, Harris WA, Pedersen RA: Inhibition of Activin/Nodal signaling promotes specification of human embryonic stem cells into neuroectoderm. Dev. Biol.313,107–117 (2008).
    • 32  Zhou J, Su P, Li D, Tsang S, Duan E, Wang F: High-efficiency induction of neural conversion in human ESCs and human induced pluripotent stem cells with a single chemical inhibitor of transforming growth factor-β superfamily receptors. Stem Cells28,1741–1750 (2010).
    • 33  Daheron L, Opitz SL, Zaehres H et al.: LIF/STAT3 signaling fails to maintain self-renewal of human embryonic stem cells. Stem Cells22,770–778 (2004).
    • 34  Ying QL, Nichols J, Chambers I, Smith A: BMP induction of Id proteins suppresses differentiation and sustains embryonic stem cell self-renewal in collaboration with STAT3. Cell115,281–292 (2003).
    • 35  Brons IG, Smithers LE, Trotter MW et al.: Derivation of pluripotent epiblast stem cells from mammalian embryos. Nature448,191–195 (2007).
    • 36  Barry F, Boynton RE, Liu B, Murphy JM: Chondrogenic differentiation of mesenchymal stem cells from bone marrow: differentiation-dependent gene expression of matrix components. Exp. Cell Res.268,189–200 (2001).
    • 37  Ng F, Boucher S, Koh S et al.: PDGF, TGF-β, and FGF signaling is important for differentiation and growth of mesenchymal stem cells (MSCs): transcriptional profiling can identify markers and signaling pathways important in differentiation of MSCs into adipogenic, chondrogenic, and osteogenic lineages. Blood112,295–307 (2008).
    • 38  Discher D, Dong C, Fredberg JJ et al.: Biomechanics: cell research and applications for the next decade. Ann. Biomed. Eng.37,847–859 (2009).
    • 39  Janmey PA, McCulloch CA: Cell mechanics: integrating cell responses to mechanical stimuli. Ann. Rev. Biomed. Eng.9,1–34 (2007).
    • 40  Hahn C, Schwartz MA: Mechanotransduction in vascular physiology and atherogenesis. Nat. Rev. Mol. Cell. Biol.10,53–62 (2009).
    • 41  Paszek MJ, Zahir N, Johnson KR et al.: Tensional homeostasis and the malignant phenotype. Cancer Cell8,241–254 (2005).
    • 42  Pelham RJ Jr, Wang Y-L: High resolution detection of mechanical forces exerted by locomoting fibroblasts on the substrate. Mol. Biol. Cell10,935–945 (1999).
    • 43  Zemel A, Rehfeldt F, Brown AE, Discher DE, Safran SA: Optimal matrix rigidity for stress fiber polarization in stem cells. Nat. Phys.6,468–473 (2010).▪▪ The alignment of non-muscle myosin (NMMII)-based stress fibers in MSCs depends non-monotonically on the matrix rigidity.
    • 44  Huebsch N, Arany PR, Mao AS et al.: Harnessing traction-mediated manipulation of the cell/matrix interface to control stem-cell fate. Nat. Mater.9,518–526 (2010).▪▪ Assessed the effects of matrix stiffness on MSCs in 3D.
    • 45  Gilbert PM, Havenstrite KL, Magnusson KE et al.: Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science329,1078–1081 (2010).▪▪ Substrate elasticity mimicking the elasticity of muscle supports self-renewal of muscle stem cells.
    • 46  Roskelley CD, Desprez PY, Bissell MJ: Extracellular matrix-dependent tissue-specific gene expression in mammary epithelial cells requires both physical and biochemical signal transduction. Proc. Natl Acad. Sci. USA91,12378–12382 (1994).
    • 47  Watt FM, Jordan PW, O’Neill CH: Cell shape controls terminal differentiation of human epidermal keratinocytes. Proc. Natl Acad. Sci. USA85,5576–5580 (1988).
    • 48  Ren XD, Kiosses WB, Schwartz MA: Regulation of the small GTP-binding protein Rho by cell adhesion and the cytoskeleton. EMBO J.18,578–585 (1999).
    • 49  Curtis A, Wilkinson C: Nantotechniques and approaches in biotechnology. Trends Biotechnol.19,97–101 (2001).
    • 50  Flemming RG, Murphy CJ, Abrams GA, Goodman SL, Nealey PF: Effects of synthetic micro- and nano-structured surfaces on cell behavior. Biomaterials20,573–588 (1999).
    • 51  Dalby MJ, Gadegaard N, Tare R et al.: The control of human mesenchymal cell differentiation using nanoscale symmetry and disorder. Nat. Mater.6,997–1003 (2007).▪▪ Investigated the effects of physical topography on MSC differentiation.
    • 52  Kurpinski K, Chu J, Hashi C, Li S: Anisotropic mechanosensing by mesenchymal stem cells. Proc. Natl Acad. Sci. USA103,16095–16100 (2006).▪▪ Mechanical strain controls MSC gene expression.
    • 53  Kurpinski K, Chu J, Wang D, Li S: Proteomic profiling of mesenchymal stem cell responses to mechanical strain and TGF-β1. Cell. Mol. Bioeng.2,606–614 (2009).▪▪ Mechanical strain and TGF-β1 co-operate to regulate MSC gene expression.
    • 54  Takahashi K, Yamanaka S: Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell126,663–676 (2006).
    • 55  Takahashi K, Tanabe K, Ohnuki M et al.: Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell131,861–872 (2007).
    • 56  Yu J, Vodyanik MA, Smuga-Otto K et al.: Induced pluripotent stem cell lines derived from human somatic cells. Science318,1917–1920 (2007).
    • 57  Park IH, Zhao R, West JA et al.: Reprogramming of human somatic cells to pluripotency with defined factors. Nature451,141–146 (2008).
    • 58  Wozniak MA, Chen CS: Mechanotransduction in development: a growing role for contractility. Nat. Rev. Mol. Cell. Biol.10,34–43 (2009).
    • 59  Chowdhury F, Na S, Li D et al.: Material properties of the cell dictate stress-induced spreading and differentiation in embryonic stem cells. Nat. Mater.9,82–88 (2010).▪▪ Adherent mouse embryonic stem cells (ESCs) are softer and much more sensitive to a local cyclic stress.
    • 60  Poh YC, Chowdhury F, Tanaka TS, Wang N: Embryonic stem cells do not stiffen on rigid substrates. Biophys. J.99,L19–L21 (2010).▪ Single ESCs do not stiffen on rigid substrates.
    • 61  Chowdhury F, Li Y, Poh YC, Yokohama-Tamaki T, Wang N, Tanaka TS: Soft substrates promote homogeneous self-renewal of embryonic stem cells via downregulating cell–matrix tractions. PLoS One5,e15655 (2010).▪ Soft substrates promote homogeneous self-renewal of mouse ESCs.
    • 62  Shimizu N, Yamamoto K, Obi S et al.: Cyclic strain induces mouse embryonic stem cell differentiation into vascular smooth muscle cells by activating PDGF receptor-β. J. Appl. Physiol.104,766–772 (2008).▪ Cyclic strain induces mouse ESC differentiation to vascular smooth muscle cells.
    • 63  Yamamoto K, Sokabe T, Watabe T et al.: Fluid shear stress induces differentiation of Flk-1-positive embryonic stem cells into vascular endothelial cells in vitro. Am. J. Physiol. Heart Circ. Physiol.288,H1915–H1924 (2005).
    • 64  Garin G, Berk BC: Flow-mediated signaling modulates endothelial cell phenotype. Endothelium13,375–384 (2006).
    • 65  Adamo L, Naveiras O, Wenzel PL et al.: Biomechanical forces promote embryonic haematopoiesis. Nature459,1131–1135 (2009).▪▪ Fluid shear stress promotes differentiation of ESC-derived hematopoietic precursors.
    • 66  Evans ND, Minelli C, Gentleman E et al.: Substrate stiffness affects early differentiation events in embryonic stem cells. Eur. Cell Mater.18,1–13; discussion 13–14 (2009).
    • 67  Peerani R, Rao BM, Bauwens C et al.: Niche-mediated control of human embryonic stem cell self-renewal and differentiation. EMBO J.26,4744–4755 (2007).▪▪ Assessed the effects of colony sizes on ESC fate.
    • 68  Ng ES, Davis RP, Azzola L, Stanley EG, Elefanty AG: Forced aggregation of defined numbers of human embryonic stem cells into embryoid bodies fosters robust, reproducible hematopoietic differentiation. Blood106,1601–1603 (2005).
    • 69  Nostro MC, Cheng X, Keller GM, Gadue P: Wnt, activin, and BMP signaling regulate distinct stages in the developmental pathway from embryonic stem cells to blood. Cell Stem Cell2,60–71 (2008).
    • 70  Friedland JC, Lee MH, Boettiger D: Mechanically activated integrin switch controls α5β1 function. Science323,642–644 (2009).
    • 71  Shin ME, He Y, Li D et al.: Spatiotemporal organization, regulation, and functions of tractions during neutrophil chemotaxis. Blood116,3297–3310 (2010).
    • 72  del Rio A, Perez-Jimenez R, Liu R, Roca-Cusachs P, Fernandez JM, Sheetz MP: Stretching single talin rod molecules activates vinculin binding. Science323,638–641 (2009).
    • 73  Grashoff C, Hoffman BD, Brenner MD et al.: Measuring mechanical tension across vinculin reveals regulation of focal adhesion dynamics. Nature466,263–266 (2010).▪ In living cells force transmission across focal adhesions is necessary for the stabilization of focal adhesions under force.
    • 74  Johnson CP, Tang HY, Carag C, Speicher DW, Discher DE: Forced unfolding of proteins within cells. Science317,663–666 (2007).▪ Described a technology to detect forced unfolding of proteins within cells.
    • 75  Lyons RM, Gentry LE, Purchio AF, Moses HL: Mechanism of activation of latent recombinant transforming growth factor-β1 by plasmin. J. Cell. Biol.110,1361–1367 (1990).
    • 76  Wipff PJ, Rifkin DB, Meister JJ, Hinz B: Myofibroblast contraction activates latent TGF-β1 from the extracellular matrix. J. Cell. Biol.179,1311–1323 (2007).▪ Myofibroblast contraction activates latent TGF-β1.
    • 77  Ahamed J, Burg N, Yoshinaga K, Janczak CA, Rifkin DB, Coller BS: In vitro and in vivo evidence for shear-induced activation of latent transforming growth factor-β1. Blood112,3650–3660 (2008).▪ Describes shear-induced activation of latent TGF-β1.
    • 78  Conti MA, Even-Ram S, Liu C, Yamada KM, Adelstein RS: Defects in cell adhesion and the visceral endoderm following ablation of nonmuscle myosin heavy chain II-A in mice. J. Biol. Chem.279,41263–41266 (2004).
    • 79  Tullio AN, Accili D, Ferrans VJ et al.: Nonmuscle myosin II-B is required for normal development of the mouse heart. Proc. Natl Acad. Sci. USA94,12407–12412 (1997).
    • 80  Watanabe K, Ueno M, Kamiya D et al.: A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat. Biotechnol.25,681–686 (2007).
    • 81  Xu Y, Zhu X, Hahm HS et al.: Revealing a core signaling regulatory mechanism for pluripotent stem cell survival and self-renewal by small molecules. Proc. Natl Acad. Sci. USA107,8129–8134 (2010).
    • 82  Li D, Zhou J, Wang L et al.: Integrated biochemical and mechanical signals regulate multifaceted human embryonic stem cell functions. J. Cell. Biol.191,631–644 (2010).▪▪ NMMIIA controls multifaceted human ESC functions.
    • 83  Chen G, Hou Z, Gulbranson DR, Thomson JA: Actin-myosin contractility is responsible for the reduced viability of dissociated human embryonic stem cells. Cell Stem Cell7,240–248 (2010).▪ Actin–myosin contractility is responsible for the reduced viability of dissociated human ESCs.
    • 84  Ohgushi M, Matsumura M, Eiraku M et al.: Molecular pathway and cell state responsible for dissociation-induced apoptosis in human pluripotent stem cells. Cell Stem Cell7,225–239 (2010).▪ The Rho–ROCK–NMMII pathway is responsible for dissociation-induced apoptosis in human ESCs.
    • 85  Shewan AM, Maddugoda M, Kraemer A et al.: Myosin 2 is a key Rho kinase target necessary for the local concentration of E-cadherin at cell–cell contacts. Mol. Biol. Cell16,4531–4542 (2005).