Nuclear Lamin-A Scales with Tissue Stiffness and Enhances Matrix-Directed Differentiation

Introduction Tissues can be soft like brain, bone marrow, and fat, which bear little mechanical stress, or stiff like muscle, cartilage, and bone, which sustain high levels of stress. Systematic relationships between tissue stiffness, protein abundance, and differential gene expression are unclear. Recent studies of stem cells cultured on matrices of different elasticity, E, have suggested that differentiation is mechanosensitive, but the molecular mechanisms involved in particular tissues remain elusive. Tissue micromechanics correlate with abundance of collagens and nuclear lamins, which influence cell differentiation. (Left) Collagen and lamin-A levels scale with E, consistent with matching tissue stress to nuclear mechanics. (Right) Matrix stiffness in tissue culture increases cell tension and stabilizes lamin-A, regulating its own transcription and that of stress fiber genes, enhancing differentiation. RA, retinoic acid, i.e., vitamin A; RARG, YAP1, and SRF, transcription factors. Methods We developed quantitative mass spectrometry algorithms to measure protein abundance, stoichiometry, conformation, and interactions within tissues and cells in relation to stiffness of tissues and extracellular matrix. Manipulations of lamin-A levels with small interfering RNA, overexpression, and retinoic acid or antagonist were applied to stem cells cultured on different matrices to assess lamin-A’s role in mechanosensitive differentiation. To characterize molecular mechanisms, promoter analyses, transcriptional profiling, and localization of transcription factors were complemented by measurements of nuclear mechanics and by modeling of the core gene circuit. Results Proteomic profiling of multiple adult solid tissues showed that widely varied levels of collagens in extracellular matrix and of lamin-A in nuclei followed power-law scaling versus E. Scaling for mechanoresponsive lamin-A conformed to predictions from polymer physics, whereas lamin-B’s varied weakly. Tumor xenograft studies further demonstrated that matrix determined tissue E, whereas lamin-A levels responded to changes in E. In tissue culture cells, both lamin-A conformation and expression were mechanosensitive, with phosphorylation and turnover of lamin-A correlating inversely with matrix E. Lamin-A knockdown enhanced mesenchymal stem cell differentiation on soft matrix that favored a low-stress, fat phenotype. Lamin-A overexpression or transcriptional induction with a retinoic acid (RA) antagonist enhanced differentiation on stiff matrix toward a high-stress, bone phenotype. Downstream of matrix stiffness, the RA pathway regulated lamin-A transcription, but feedback by lamin-A regulated RA receptor (RARG) translocation into nuclei. High lamin-A levels physically impeded nuclear remodeling under stress but also coregulated other key factors. These factors included both serum response factor (SRF), which promoted expression of stress fiber–associated proteins involved in differentiation, and a Hippo pathway factor (YAP1) involved in growth. Discussion The characteristic stress in normal tissue favors collagen accumulation and a characteristic stiffness that cells transduce through nuclear lamin-A to enhance tissue-specific differentiation. Tension-inhibited turnover of rope-like filaments of lamin-A provides sufficient mechanochemical control of a core gene circuit to explain the steady-state scaling of lamin-A with E. High lamin-A physically stabilizes the nucleus against stress and thereby stabilizes the nuclear lamina and chromatin, with implications for epigenetic stabilization and limiting of DNA breaks. Moreover, lamin-A levels directly or indirectly regulate many proteins involved in tissue-specific gene expression, and, because lamin-A levels can vary by a factor of 10 or more downstream of tissue mechanics, an important fraction of tissue-specific gene expression depends on tissue mechanics, which changes in development, injury, and many diseases. Lamins and Tissue Stiffness Microenvironment can influence cell fate and behavior; for example, extracellular matrix (ECM) stiffness increases cell proliferation, and ECM rigidity induces disorders in tissue morphogenesis by increasing cell tension. Swift et al. (1240104; see the Perspective by Bainer and Weaver) used proteomics to identify molecules that are mechanical sensors for tissue elasticity in the nucleus and discovered that expression of lamin-A levels apparently functions as a “mechanostat.” Tissues that need to remain stiff under stress rely on lamin-A to keep the cell nucleus whole. [Also see Perspective by Bainer and Weaver] Tissues can be soft like fat, which bears little stress, or stiff like bone, which sustains high stress, but whether there is a systematic relationship between tissue mechanics and differentiation is unknown. Here, proteomics analyses revealed that levels of the nucleoskeletal protein lamin-A scaled with tissue elasticity, E, as did levels of collagens in the extracellular matrix that determine E. Stem cell differentiation into fat on soft matrix was enhanced by low lamin-A levels, whereas differentiation into bone on stiff matrix was enhanced by high lamin-A levels. Matrix stiffness directly influenced lamin-A protein levels, and, although lamin-A transcription was regulated by the vitamin A/retinoic acid (RA) pathway with broad roles in development, nuclear entry of RA receptors was modulated by lamin-A protein. Tissue stiffness and stress thus increase lamin-A levels, which stabilize the nucleus while also contributing to lineage determination.

[1]  Adam J. Engler,et al.  Myotubes differentiate optimally on substrates with tissue-like stiffness , 2004, The Journal of cell biology.

[2]  Wutian Wu,et al.  Implantation of adult bone marrow-derived mesenchymal stem cells transfected with the neurotrophin-3 gene and pretreated with retinoic acid in completely transected spinal cord , 2010, Brain Research.

[3]  K. Wilson,et al.  The nucleoskeleton as a genome-associated dynamic 'network of networks' , 2011, Nature Reviews Molecular Cell Biology.

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

[5]  N. Maraldi,et al.  The protein kinase Akt/PKB regulates both prelamin A degradation and Lmna gene expression , 2013, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[6]  Wolfgang Schütz,et al.  A truncated lamin A in the Lmna−/− mouse line , 2012, Nucleus.

[7]  P. Lu,et al.  Serum Response Factor Is Required for Cortical Axon Growth But Is Dispensable for Neurogenesis and Neocortical Lamination , 2011, The Journal of Neuroscience.

[8]  C. Grund,et al.  Rearrangement of the vimentin cytoskeleton during adipose conversion: Formation of an intermediate filament cage around lipid globules , 1987, Cell.

[9]  E. Olson,et al.  A WW domain protein TAZ is a critical coactivator for TBX5, a transcription factor implicated in Holt-Oram syndrome. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[10]  S. K. Zaidi,et al.  Tyrosine phosphorylation controls Runx2‐mediated subnuclear targeting of YAP to repress transcription , 2004, The EMBO journal.

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

[12]  D. Discher,et al.  Power-law rheology of isolated nuclei with deformation mapping of nuclear substructures. , 2005, Biophysical journal.

[13]  J. Hope Stereological analysis of the ultrastructure of liver parenchymal cells during pregnancy and lactation. , 1970, Journal of ultrastructure research.

[14]  Adam J. Engler,et al.  Mesenchymal stem cell injection after myocardial infarction improves myocardial compliance , 2006 .

[15]  S. Shoelson,et al.  Structure of the Globular Tail of Nuclear Lamin* , 2002, The Journal of Biological Chemistry.

[16]  Shuiliang Yu,et al.  Retinoic Acid Induces Neurogenesis by Activating Both Retinoic Acid Receptors (RARs) and Peroxisome Proliferator-activated Receptor β/δ (PPARβ/δ)* , 2012, The Journal of Biological Chemistry.

[17]  H. Drissi,et al.  Effect of Lamin A/C Knockdown on Osteoblast Differentiation and Function , 2009, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[18]  K. Okumura,et al.  c-Jun and Sp1 family are critical for retinoic acid induction of the lamin A/C retinoic acid-responsive element. , 2004, Biochemical and biophysical research communications.

[19]  B. Chen,et al.  DNA microarray analysis of gene expression in endothelial cells in response to 24-h shear stress. , 2001, Physiological genomics.

[20]  William White,et al.  A Proposal , 2008, Moon, Sun, and Witches.

[21]  D. Ingber,et al.  Mechanotransduction at a distance: mechanically coupling the extracellular matrix with the nucleus , 2009, Nature Reviews Molecular Cell Biology.

[22]  J. Lammerding,et al.  Lamin A/C and emerin regulate MKL1/SRF activity by modulating actin dynamics , 2013, Nature.

[23]  P. Janmey,et al.  Nonlinear elasticity in biological gels , 2004, Nature.

[24]  Ueli Aebi,et al.  Early detection of aging cartilage and osteoarthritis in mice and patient samples using atomic force microscopy. , 2009, Nature nanotechnology.

[25]  T. Misteli,et al.  Lamin A-dependent misregulation of adult stem cells associated with accelerated ageing , 2008, Nature Cell Biology.

[26]  T. Underhill,et al.  The retinoic acid binding protein CRABP2 is increased in murine models of degenerative joint disease , 2009, Arthritis research & therapy.

[27]  Roger D. Kamm,et al.  Computational Analysis of Viscoelastic Properties of Crosslinked Actin Networks , 2009, PLoS Comput. Biol..

[28]  K. Beck,et al.  Supercoiled Protein Motifs: The Collagen Triple-Helix and the α-Helical Coiled Coil , 1998 .

[29]  U. Aebi,et al.  Intermediate filaments: primary determinants of cell architecture and plasticity. , 2009, The Journal of clinical investigation.

[30]  K. Moore,et al.  Stem Cells and Their Niches , 2006, Science.

[31]  S. Lees-Miller,et al.  Repair of ionizing radiation-induced DNA double-strand breaks by non-homologous end-joining. , 2009, The Biochemical journal.

[32]  K. Wilson,et al.  Direct actin binding to A- and B-type lamin tails and actin filament bundling by the lamin A tail , 2010, Nucleus.

[33]  Thomas Werner,et al.  MatInspector and beyond: promoter analysis based on transcription factor binding sites , 2005, Bioinform..

[34]  R. Goldman,et al.  Nuclear lamins: building blocks of nuclear architecture. , 2002, Genes & development.

[35]  F. Hosoda,et al.  A BAC-based STS-content map spanning a 35-Mb region of human chromosome 1p35-p36. , 2001, Genomics.

[36]  D. Discher,et al.  Crawling from soft to stiff matrix polarizes the cytoskeleton and phosphoregulates myosin-II heavy chain , 2012, The Journal of cell biology.

[37]  《中华放射肿瘤学杂志》编辑部 Medline , 2001, Current Biology.

[38]  Richard T. Lee,et al.  Lamins A and C but Not Lamin B1 Regulate Nuclear Mechanics* , 2006, Journal of Biological Chemistry.

[39]  M. Layne,et al.  Myocardin-related Transcription Factor-A Complexes Activate Type I Collagen Expression in Lung Fibroblasts* , 2011, The Journal of Biological Chemistry.

[40]  Bradley E. Bernstein,et al.  DNA Sequence-Dependent Compartmentalization and Silencing of Chromatin at the Nuclear Lamina , 2012, Cell.

[41]  D. Discher,et al.  Myosin-II inhibition and soft 2D matrix maximize multinucleation and cellular projections typical of platelet-producing megakaryocytes , 2011, Proceedings of the National Academy of Sciences.

[42]  Dennis E Discher,et al.  Minimal " Self " Peptides That Inhibit Phagocytic Clearance and Enhance Delivery of Nanoparticles References and Notes , 2022 .

[43]  Juliet A. Ellis,et al.  Mammalian SUN Protein Interaction Networks at the Inner Nuclear Membrane and Their Role in Laminopathy Disease Processes* , 2009, The Journal of Biological Chemistry.

[44]  N. Lane,et al.  Glucocorticoid excess in mice results in early activation of osteoclastogenesis and adipogenesis and prolonged suppression of osteogenesis: a longitudinal study of gene expression in bone tissue from glucocorticoid-treated mice. , 2008, Arthritis and rheumatism.

[45]  S. Zinn-Justin,et al.  The Ig-like structure of the C-terminal domain of lamin A/C, mutated in muscular dystrophies, cardiomyopathy, and partial lipodystrophy. , 2002, Structure.

[46]  T. Tuschl,et al.  Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells , 2001, Nature.

[47]  D. Vaux,et al.  The apparent absence of lamin B1 and emerin in many tissue nuclei is due to epitope masking , 2005, Journal of Molecular Histology.

[48]  B. Chen,et al.  DNA microarray analysis of gene expression in endothelial cells in response to 24-h shear stress. , 2001, Physiological genomics.

[49]  Li Li,et al.  Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. , 2007, Genes & development.

[50]  F. McKeon,et al.  Mutations of phosphorylation sites in lamin A that prevent nuclear lamina disassembly in mitosis , 1990, Cell.

[51]  N. Maraldi,et al.  Lamin A Ser404 is a nuclear target of Akt phosphorylation in C2C12 cells. , 2008, Journal of proteome research.

[52]  Roberto Bolli,et al.  Life and Death of Cardiac Stem Cells: A Paradigm Shift in Cardiac Biology , 2006, Circulation.

[53]  A. Sharov,et al.  Mouse B-Type Lamins Are Required for Proper Organogenesis But Not by Embryonic Stem Cells , 2011, Science.

[54]  Wesley R. Legant,et al.  Degradation-mediated cellular traction directs stem cell fate in covalently crosslinked three-dimensional hydrogels , 2013, Nature materials.

[55]  D. E. Olins,et al.  Nuclear envelope and chromatin compositional differences comparing undifferentiated and retinoic acid- and phorbol ester-treated HL-60 cells. , 2001, Experimental cell research.

[56]  S. Young,et al.  Regulation of prelamin A but not lamin C by miR-9, a brain-specific microRNA , 2012, Proceedings of the National Academy of Sciences.

[57]  Farshid Guilak,et al.  Zonal Uniformity in Mechanical Properties of the Chondrocyte Pericellular Matrix: Micropipette Aspiration of Canine Chondrons Isolated by Cartilage Homogenization , 2005, Annals of Biomedical Engineering.

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

[59]  P. Janmey,et al.  Increased stiffness of the rat liver precedes matrix deposition: implications for fibrosis. , 2007, American journal of physiology. Gastrointestinal and liver physiology.

[60]  J. Chamberlain,et al.  Dystrophin‐deficient mdx mice display a reduced life span and are susceptible to spontaneous rhabdomyosarcoma , 2007, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[61]  P. Wieacker,et al.  The retinol acid receptor B gene is hypermethylated in patients with familial partial lipodystrophy. , 2007, Journal of molecular endocrinology.

[62]  Roland Rad,et al.  Rapid and efficient reprogramming of somatic cells to induced pluripotent stem cells by retinoic acid receptor gamma and liver receptor homolog 1 , 2011, Proceedings of the National Academy of Sciences.

[63]  G. Strobl,et al.  The Physics of Polymers: Concepts for Understanding their Structures and Behavior ~Springer , 1997 .

[64]  C. Glover,et al.  Gene expression profiling for hematopoietic cell culture , 2006 .

[65]  R. Hyatt,et al.  Effects of age on elastic moduli of human lungs. , 2000, Journal of applied physiology.

[66]  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.

[67]  R. Eckert,et al.  AGN193109 Is a Highly Effective Antagonist of Retinoid Action in Human Ectocervical Epithelial Cells (*) , 1996, The Journal of Biological Chemistry.

[68]  Dennis E Discher,et al.  How deeply cells feel: methods for thin gels , 2010, Journal of physics. Condensed matter : an Institute of Physics journal.

[69]  J. Ayala-Sumuano,et al.  Srebf1a is a key regulator of transcriptional control for adipogenesis , 2011, Scientific reports.

[70]  Kaushik Sengupta,et al.  Nuclear lamins: major factors in the structural organization and function of the nucleus and chromatin. , 2008, Genes & development.

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

[72]  Eben Alsberg,et al.  FRET measurements of cell-traction forces and nano-scale clustering of adhesion ligands varied by substrate stiffness. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[73]  David A Weitz,et al.  Biophysical properties of normal and diseased renal glomeruli. , 2011, American journal of physiology. Cell physiology.

[74]  Hui-Ann Wu,et al.  Serum response factor regulates bone formation via IGF‐1 and Runx2 signals , 2012, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[75]  Xi Ma,et al.  SiRNA against Fabp5 induces 3T3-L1 cells apoptosis during adipocytic induction , 2010, Molecular Biology Reports.

[76]  F. Smedts,et al.  A- and B-type lamins are differentially expressed in normal human tissues , 1997, Histochemistry and Cell Biology.

[77]  E. Olson,et al.  Requirement of a Myocardin-Related Transcription Factor for Development of Mammary Myoepithelial Cells , 2006, Molecular and Cellular Biology.

[78]  I. Nonaka,et al.  Human PTRF mutations cause secondary deficiency of caveolins resulting in muscular dystrophy with generalized lipodystrophy. , 2009, The Journal of clinical investigation.

[79]  Eran Meshorer,et al.  Global epigenetic changes during somatic cell reprogramming to iPS cells. , 2011, Journal of molecular cell biology.

[80]  M. Hetzer,et al.  MEL‐28/ELYS is required for the recruitment of nucleoporins to chromatin and postmitotic nuclear pore complex assembly , 2007, EMBO reports.

[81]  P. Janmey,et al.  Bone marrow-derived human mesenchymal stem cells become quiescent on soft substrates but remain responsive to chemical or mechanical stimuli. , 2009, Tissue engineering. Part A.

[82]  S. Thrun,et al.  Substrate Elasticity Regulates Skeletal Muscle Stem Cell Self-Renewal in Culture , 2010, Science.

[83]  David F Meaney,et al.  Matrices with compliance comparable to that of brain tissue select neuronal over glial growth in mixed cortical cultures. , 2006, Biophysical journal.

[84]  H. Frost Bone “mass” and the “mechanostat”: A proposal , 1987, The Anatomical record.

[85]  Hong Zhang,et al.  Targeting multiple signal transduction pathways through inhibition of Hsp90 , 2004, Journal of Molecular Medicine.

[86]  M. Nelson,et al.  Nuclear Factor of Activated T Cells and Serum Response Factor Cooperatively Regulate the Activity of an α-Actin Intronic Enhancer* , 2005, Journal of Biological Chemistry.

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

[88]  W. D. De Vos,et al.  Repetitive disruptions of the nuclear envelope invoke temporary loss of cellular compartmentalization in laminopathies. , 2011, Human molecular genetics.

[89]  A. Cumano,et al.  Forced Unfolding of Proteins Within Cells , 2007 .

[90]  C. Chapman,et al.  Enhancing the Efficacy of Drug-loaded Nanocarriers against Brain Tumors by Targeted Radiation Therapy , 2013, Oncotarget.

[91]  Brian Burke,et al.  Loss of a-Type Lamin Expression Compromises Nuclear Envelope Integrity Leading to Muscular Dystrophy , 1999, The Journal of cell biology.

[92]  Bin Zhao,et al.  The Hippo pathway in organ size control, tissue regeneration and stem cell self-renewal , 2011, Nature Cell Biology.

[93]  D. G. T. Strange,et al.  Extracellular-matrix tethering regulates stem-cell fate. , 2012, Nature materials.

[94]  T. Misteli,et al.  Identification of differential protein interactors of lamin A and progerin , 2010, Nucleus.

[95]  G. Shivashankar Mechanosignaling to the cell nucleus and gene regulation. , 2011, Annual review of biophysics.

[96]  D. Discher,et al.  Mechanobiology of bone marrow stem cells: from myosin-II forces to compliance of matrix and nucleus in cell forms and fates. , 2013, Differentiation; research in biological diversity.

[97]  Jennifer L West,et al.  Poly(ethylene glycol) hydrogel system supports preadipocyte viability, adhesion, and proliferation. , 2005, Tissue engineering.

[98]  Dennis E. Discher,et al.  Physical plasticity of the nucleus in stem cell differentiation , 2007, Proceedings of the National Academy of Sciences.

[99]  Manolis Kellis,et al.  Constitutive nuclear lamina–genome interactions are highly conserved and associated with A/T-rich sequence , 2013, Genome research.

[100]  Adam J Engler,et al.  Embryonic cardiomyocytes beat best on a matrix with heart-like elasticity: scar-like rigidity inhibits beating , 2008, Journal of Cell Science.

[101]  P. Chambon,et al.  All-trans and 9-cis retinoic acid induction of CRABPII transcription is mediated by RAR-RXR heterodimers bound to DR1 and DR2 repeated motifs , 1992, Cell.

[102]  Charles A. DiMarzio,et al.  Mechanical Strain Stabilizes Reconstituted Collagen Fibrils against Enzymatic Degradation by Mammalian Collagenase Matrix Metalloproteinase 8 (MMP-8) , 2010, PloS one.

[103]  Michael P. Sheetz,et al.  Force Sensing by Mechanical Extension of the Src Family Kinase Substrate p130Cas , 2006, Cell.

[104]  R. Grosse,et al.  Nuclear Actin Network Assembly by Formins Regulates the SRF Coactivator MAL , 2013, Science.

[105]  J. Winkler,et al.  Analysis of neurogenesis and programmed cell death reveals a self-renewing capacity in the adult rat brain , 2000, Neuroscience Letters.

[106]  Thomas Cremer,et al.  The A- and B-type nuclear lamin networks: microdomains involved in chromatin organization and transcription. , 2008, Genes & development.

[107]  Thomas Benjamin,et al.  TAZ, a Transcriptional Modulator of Mesenchymal Stem Cell Differentiation , 2005, Science.

[108]  L. Kaufman,et al.  Elastic moduli of collagen gels can be predicted from two-dimensional confocal microscopy. , 2009, Biophysical journal.

[109]  H. Worman Nuclear lamins and laminopathies , 2012, The Journal of pathology.

[110]  S. Adam,et al.  Functions and dysfunctions of the nuclear lamin Ig-fold domain in nuclear assembly, growth, and Emery–Dreifuss muscular dystrophy , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[111]  R. Kosfeld,et al.  The Physics of Polymers, Concepts for Understanding Their Structures and Behavior , 1998 .

[112]  Alfred Nordheim,et al.  Linking actin dynamics and gene transcription to drive cellular motile functions , 2010, Nature Reviews Molecular Cell Biology.

[113]  Ronald G. Larson,et al.  The scaling of zero-shear viscosities of semidilute polymer solutions with concentration , 2005 .

[114]  R. Rosenfeld Nature , 2009, Otolaryngology--head and neck surgery : official journal of American Academy of Otolaryngology-Head and Neck Surgery.

[115]  Jiandie D. Lin,et al.  TEAD mediates YAP-dependent gene induction and growth control. , 2008, Genes & development.

[116]  Jungho Kim,et al.  Pyruvate kinase isozyme type M2 (PKM2) interacts and cooperates with Oct-4 in regulating transcription. , 2008, The international journal of biochemistry & cell biology.

[117]  Sanjay Kumar,et al.  The mechanical rigidity of the extracellular matrix regulates the structure, motility, and proliferation of glioma cells. , 2009, Cancer research.

[118]  K. Beck,et al.  Supercoiled protein motifs: the collagen triple-helix and the alpha-helical coiled coil. , 1998, Journal of structural biology.

[119]  M. Sheetz,et al.  Local force and geometry sensing regulate cell functions , 2006, Nature Reviews Molecular Cell Biology.

[120]  M. Eijken,et al.  Unraveling the human bone microenvironment beyond the classical extracellular matrix proteins: a human bone protein library. , 2011, Journal of proteome research.

[121]  David Haussler,et al.  ENCODE Data in the UCSC Genome Browser: year 5 update , 2012, Nucleic Acids Res..

[122]  Masahiro Iwamoto,et al.  Potent Inhibition of Heterotopic Ossification by Nuclear Retinoic Acid Receptor γ Agonists , 2011, Nature Medicine.

[123]  F. Watt,et al.  Actin and serum response factor transduce physical cues from the microenvironment to regulate epidermal stem cell fate decisions , 2010, Nature Cell Biology.

[124]  D. E. Discher,et al.  Matrix elasticity directs stem cell lineage — Soluble factors that limit osteogenesis , 2009 .

[125]  R. Trembath,et al.  A novel interaction between lamin A and SREBP1: implications for partial lipodystrophy and other laminopathies. , 2002, Human molecular genetics.

[126]  W. Gardner,et al.  Carcinogenesis , 1961, The Yale Journal of Biology and Medicine.

[127]  T. C. B. McLeish,et al.  Polymer Physics , 2009, Encyclopedia of Complexity and Systems Science.

[128]  L. Gudas,et al.  Gene expression profiling elucidates a specific role for RARgamma in the retinoic acid-induced differentiation of F9 teratocarcinoma stem cells. , 2008, Biochemical pharmacology.