451
|
Denisin AK, Pruitt BL. Tuning the Range of Polyacrylamide Gel Stiffness for Mechanobiology Applications. ACS APPLIED MATERIALS & INTERFACES 2016; 8:21893-21902. [PMID: 26816386 DOI: 10.1021/acsami.5b09344] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Adjusting the acrylamide monomer and cross-linker content in polyacrylamide gels controls the hydrogel stiffness, yet the reported elastic modulus for the same formulations varies widely and these discrepancies are frequently attributed to different measurement methods. Few studies exist that examine stiffness trends across monomer and cross-linker concentrations using the same characterization platform. In this work, we use Atomic Force Microscopy and analyze force-distance curves to derive the elastic modulus of polyacrylamide hydrogels. We find that gel elastic modulus increases with increasing cross-link concentration until an inflection point, after which gel stiffness decreases with increasing cross-linking. This behavior arises because of the formation of highly cross-linked clusters, which add inhomogeneity and heterogeneity to the network structure, causing the global network to soften even under high cross-linking conditions. We identify these inflection points for three different total polymer formulations. When we alter gelation kinetics by using a low polymerization temperature, we find that gels are stiffer when polymerized at 4 °C compared to room temperature, indicating a complex relationship between gel structure, elasticity, and network formation. We also investigate how gel stiffness changes during storage over 10 days and find that specific gel formulations undergo significant stiffening (1.55 ± 0.13), which may be explained by differences in gel swelling resulting from initial polymerization parameters. Taken together, our study emphasizes the importance of polyacrylamide formulation, polymerization temperature, gelation time, and storage duration in defining the structural and mechanical properties of the polyacrylamide hydrogels.
Collapse
Affiliation(s)
- Aleksandra K Denisin
- Department of Bioengineering, Stanford University , 443 Via Ortega, Shriram Center, Room 119, Stanford, California 94305-4125, United States
- Department of Mechanical Engineering, Stanford University , Building 530 440 Escondido Mall, Stanford, California 94305, United States
| | - Beth L Pruitt
- Department of Mechanical Engineering, Stanford University , Building 530 440 Escondido Mall, Stanford, California 94305, United States
- Stanford Cardiovascular Institute, Stanford University , 265 Campus Drive Stanford, California 94305, United States
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine , 279 Campus Drive, Beckman Center, Room B100A, Stanford, California 94305-5345, United States
| |
Collapse
|
452
|
Sheikhi A, Hill RJ. Hydrogel-colloid interfacial interactions: a study of tailored adhesion using optical tweezers. SOFT MATTER 2016; 12:6575-6587. [PMID: 27425660 DOI: 10.1039/c6sm00903d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Dynamics of colloidal particles adhering to soft, deformable substrates, such as tissues, biofilms, and hydrogels play a key role in many biological and biomimetic processes. These processes, including, but not limited to colloid-based delivery, stitching, and sorting, involve microspheres exploring the vicinity of soft, sticky materials in which the colloidal dynamics are affected by the fluid environment (e.g., viscous coupling), inter-molecular interactions between the colloids and substrates (e.g., Derjaguin-Landau-Verwey-Overbeek (DLVO) theory), and the viscoelastic properties of contact region. To better understand colloidal dynamics at soft interfaces, an optical tweezers back-focal-plane interferometry apparatus was developed to register the transverse Brownian motion of a silica microsphere in the vicinity of polyacrylamide (PA) hydrogel films. The time-dependent mean-squared displacements are well described by a single exponential relaxation, furnishing measures of the transverse interfacial diffusion coefficient and binding stiffness. Substrates with different elasticities were prepared by changing the PA crosslinking density, and the inter-molecular interactions were adjusted by coating the microspheres with fluid membranes. Stiffer PA hydrogels (with bulk Young's moduli ≈1-10 kPa) immobilize the microspheres more firmly (lower diffusion coefficient and position variance), and coating the particles with zwitterionic lipid bilayers (DOPC) completely eliminates adhesion, possibly by repulsive dispersion forces. Remarkably, embedding polyethylene glycol-grafted lipid bilayers (DSPE-PEG2k-Amine) in the zwitterionic fluid membranes produces stronger adhesion, possibly because of polymer-hydrogel attraction and entanglement. This study provides new insights to guide the design of nanoparticles and substrates with tunable adhesion, leading to smarter delivery, sorting, and screening of micro- and nano-systems.
Collapse
Affiliation(s)
- Amir Sheikhi
- Department of Chemical Engineering, McGill University, 3610 University Street, Montreal, Quebec H3A 0C5, Canada.
| | | |
Collapse
|
453
|
Kourouklis AP, Kaylan KB, Underhill GH. Substrate stiffness and matrix composition coordinately control the differentiation of liver progenitor cells. Biomaterials 2016; 99:82-94. [DOI: 10.1016/j.biomaterials.2016.05.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/30/2016] [Accepted: 05/11/2016] [Indexed: 02/07/2023]
|
454
|
Lee J, Abdeen AA, Wycislo KL, Fan TM, Kilian KA. Interfacial geometry dictates cancer cell tumorigenicity. NATURE MATERIALS 2016; 15:856-62. [PMID: 27043781 DOI: 10.1038/nmat4610] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 02/25/2016] [Indexed: 05/26/2023]
Abstract
Within the heterogeneous architecture of tumour tissue there exists an elusive population of stem-like cells that are implicated in both recurrence and metastasis. Here, by using engineered extracellular matrices, we show that geometric features at the perimeter of tumour tissue will prime a population of cells with a stem-cell-like phenotype. These cells show characteristics of cancer stem cells in vitro, as well as enhanced tumorigenicity in murine models of primary tumour growth and pulmonary metastases. We also show that interfacial geometry modulates cell shape, adhesion through integrin α5β1, MAPK and STAT activity, and initiation of pluripotency signalling. Our results for several human cancer cell lines suggest that interfacial geometry triggers a general mechanism for the regulation of cancer-cell state. Similar to how a growing tumour can co-opt normal soluble signalling pathways, our findings demonstrate how cancer can also exploit geometry to orchestrate oncogenesis.
Collapse
Affiliation(s)
- Junmin Lee
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Amr A Abdeen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Kathryn L Wycislo
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Kristopher A Kilian
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| |
Collapse
|
455
|
Li J, Mkrtschjan MA, Lin YH, Russell B. Variation in stiffness regulates cardiac myocyte hypertrophy via signaling pathways. Can J Physiol Pharmacol 2016; 94:1178-1186. [PMID: 27486838 DOI: 10.1139/cjpp-2015-0578] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Much diseased human myocardial tissue is fibrotic and stiff, which increases the work that the ventricular myocytes must perform to maintain cardiac output. The hypothesis tested is that the increased load due to greater stiffness of the substrata drives sarcomere assembly of cells, thus strengthening them. Neonatal rat ventricular myocytes (NRVM) were cultured on polyacrylamide or polydimethylsiloxane substrates with stiffness of 10 kPa, 100 kPa, or 400 kPa, or glass with stiffness of 61.9 GPa. Cell size increased with stiffness. Two signaling pathways were explored, phosphorylation of focal adhesion kinase (p-FAK) and lipids by phosphatidylinositol 4,5-bisphosphate (PIP2). Subcellular distributions of both were determined in the sarcomeric fraction by antibody localization, and total amounts were measured by Western or dot blotting, respectively. More p-FAK and PIP2 distributed to the sarcomeres of NRVM grown on stiffer substrates. Actin assembly involves the actin capping protein Z (CapZ). Both actin and CapZ dynamic exchange were significantly increased on stiffer substrates when assessed by fluorescence recovery after photobleaching (FRAP) of green fluorescent protein tags. Blunting of actin FRAP by FAK inhibition implicates linkage from mechano-signalling pathways to cell growth. Thus, increased stiffness of cardiac disease can be modeled with polymeric materials to understand how the microenvironment regulates cardiac hypertrophy.
Collapse
Affiliation(s)
- Jieli Li
- a Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave, Chicago, IL 60612, USA
| | - Michael A Mkrtschjan
- b Department of Bioengineering, University of Illinois at Chicago, 851 South Morgan Street, Chicago, IL 60607, USA
| | - Ying-Hsi Lin
- a Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave, Chicago, IL 60612, USA
| | - Brenda Russell
- a Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, 835 S. Wolcott Ave, Chicago, IL 60612, USA.,b Department of Bioengineering, University of Illinois at Chicago, 851 South Morgan Street, Chicago, IL 60607, USA
| |
Collapse
|
456
|
Lin HH, Lin HK, Lin IH, Chiou YW, Chen HW, Liu CY, Harn HIC, Chiu WT, Wang YK, Shen MR, Tang MJ. Mechanical phenotype of cancer cells: cell softening and loss of stiffness sensing. Oncotarget 2016; 6:20946-58. [PMID: 26189182 PMCID: PMC4673241 DOI: 10.18632/oncotarget.4173] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/02/2015] [Indexed: 01/06/2023] Open
Abstract
The stiffness sensing ability is required to respond to the stiffness of the matrix. Here we determined whether normal cells and cancer cells display distinct mechanical phenotypes. Cancer cells were softer than their normal counterparts, regardless of the type of cancer (breast, bladder, cervix, pancreas, or Ha-RasV12-transformed cells). When cultured on matrices of varying stiffness, low stiffness decreased proliferation in normal cells, while cancer cells and transformed cells lost this response. Thus, cancer cells undergo a change in their mechanical phenotype that includes cell softening and loss of stiffness sensing. Caveolin-1, which is suppressed in many tumor cells and in oncogene-transformed cells, regulates the mechanical phenotype. Caveolin-1-upregulated RhoA activity and Y397FAK phosphorylation directed actin cap formation, which was positively correlated with cell elasticity and stiffness sensing in fibroblasts. Ha-RasV12-induced transformation and changes in the mechanical phenotypes were reversed by re-expression of caveolin-1 and mimicked by the suppression of caveolin-1 in normal fibroblasts. This is the first study to describe this novel role for caveolin-1, linking mechanical phenotype to cell transformation. Furthermore, mechanical characteristics may serve as biomarkers for cell transformation.
Collapse
Affiliation(s)
- Hsi-Hui Lin
- Department of Physiology, National Cheng Kung University, Tainan, Taiwan
| | - Hsiu-Kuan Lin
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| | - I-Hsuan Lin
- Department of Physiology, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Wei Chiou
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Horn-Wei Chen
- Department of Physiology, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Yi Liu
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Hans I-Chen Harn
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Yang-Kao Wang
- Department of Cell Biology and Anatomy, National Cheng Kung University, Tainan, Taiwan
| | - Meng-Ru Shen
- Department of Pharmacology, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Jer Tang
- Department of Physiology, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
457
|
YAP and TAZ control peripheral myelination and the expression of laminin receptors in Schwann cells. Nat Neurosci 2016; 19:879-87. [PMID: 27273766 PMCID: PMC4925303 DOI: 10.1038/nn.4316] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 05/04/2016] [Indexed: 12/13/2022]
Abstract
Myelination is essential for nervous system function. Schwann cells interact with neurons and the basal lamina to myelinate axons, using known receptors, signals and transcription factors. In contrast, the transcriptional control of axonal sorting and the role of mechanotransduction in myelination are largely unknown. Yap and Taz are effectors of the Hippo pathway that integrate chemical and mechanical signals in cells. We describe a previously unknown role for the Hippo pathway in myelination. Using conditional mutagenesis in mice we show that Taz is required in Schwann cells for radial sorting and myelination, and that Yap is redundant with Taz. Yap/Taz are activated in Schwann cells by mechanical stimuli, and regulate Schwann cell proliferation and transcription of basal lamina receptor genes, both necessary for proper radial sorting of axons and subsequent myelination. These data link transcriptional effectors of the Hippo pathway and of mechanotransduction to myelin formation in Schwann cells.
Collapse
|
458
|
Nam S, Hu KH, Butte MJ, Chaudhuri O. Strain-enhanced stress relaxation impacts nonlinear elasticity in collagen gels. Proc Natl Acad Sci U S A 2016; 113:5492-7. [PMID: 27140623 PMCID: PMC4878492 DOI: 10.1073/pnas.1523906113] [Citation(s) in RCA: 197] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The extracellular matrix (ECM) is a complex assembly of structural proteins that provides physical support and biochemical signaling to cells in tissues. The mechanical properties of the ECM have been found to play a key role in regulating cell behaviors such as differentiation and malignancy. Gels formed from ECM protein biopolymers such as collagen or fibrin are commonly used for 3D cell culture models of tissue. One of the most striking features of these gels is that they exhibit nonlinear elasticity, undergoing strain stiffening. However, these gels are also viscoelastic and exhibit stress relaxation, with the resistance of the gel to a deformation relaxing over time. Recent studies have suggested that cells sense and respond to both nonlinear elasticity and viscoelasticity of ECM, yet little is known about the connection between nonlinear elasticity and viscoelasticity. Here, we report that, as strain is increased, not only do biopolymer gels stiffen but they also exhibit faster stress relaxation, reducing the timescale over which elastic energy is dissipated. This effect is not universal to all biological gels and is mediated through weak cross-links. Mechanistically, computational modeling and atomic force microscopy (AFM) indicate that strain-enhanced stress relaxation of collagen gels arises from force-dependent unbinding of weak bonds between collagen fibers. The broader effect of strain-enhanced stress relaxation is to rapidly diminish strain stiffening over time. These results reveal the interplay between nonlinear elasticity and viscoelasticity in collagen gels, and highlight the complexity of the ECM mechanics that are likely sensed through cellular mechanotransduction.
Collapse
Affiliation(s)
- Sungmin Nam
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305
| | - Kenneth H Hu
- Biophysics Program, Stanford University, Stanford, CA 94305
| | - Manish J Butte
- Department of Pediatrics, Stanford University, Stanford, CA 94305
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305;
| |
Collapse
|
459
|
Chaudhry A, Yazdi IK, Kongari R, Tasciotti E, Righetti R. A New Class of Phantom Materials for Poroelastography Imaging Techniques. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:1230-8. [PMID: 26806439 DOI: 10.1016/j.ultrasmedbio.2015.12.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/17/2015] [Accepted: 12/14/2015] [Indexed: 05/20/2023]
Abstract
Poroelastography is an elastographic technique used to image the temporal mechanical behavior of tissues. One of the major challenges in determining experimental potentials and limitations of this technique has been the lack of complex and realistic controlled phantoms that could be used to corroborate the limited number of theoretical and simulation studies available in the literature as well as to predict its performance in complex experimental situations and in a variety of conditions. In the study described here, we propose and analyze a new class of phantom materials for temporal elastography imaging. The results indicate that, by using polyacrylamide, we can generate inhomogeneous elastographic phantoms with controlled fluid content and fluid flow properties, while maintaining mechanical and ultrasonic properties similar to those of soft tissues.
Collapse
Affiliation(s)
- Anuj Chaudhry
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, Texas, USA
| | - Iman K Yazdi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA; Department of Biomedical Engineering, University of Houston, Houston, Texas, USA
| | - Rohit Kongari
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Ennio Tasciotti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Raffaella Righetti
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, Texas, USA.
| |
Collapse
|
460
|
Canver AC, Ngo O, Urbano RL, Clyne AM. Endothelial directed collective migration depends on substrate stiffness via localized myosin contractility and cell-matrix interactions. J Biomech 2016; 49:1369-1380. [DOI: 10.1016/j.jbiomech.2015.12.037] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 11/25/2015] [Accepted: 12/16/2015] [Indexed: 01/05/2023]
|
461
|
Abstract
There is growing appreciation of the role that the extracellular environment plays in regulating cell behavior. Mechanical, structural, and compositional cues, either alone or in concert, can drastically alter cell function. Biomaterials, and particularly hydrogels, have been developed and implemented to present defined subsets of these cues for investigating countless cellular processes as a means of understanding morphogenesis, aging, and disease. Although most scientists concede that standard cell culture materials (tissue culture plastic and glass) do a poor job of recapitulating native cellular milieus, there is currently a knowledge barrier for many researchers in regard to the application of hydrogels for cell culture. Here, we introduce hydrogels to those who may be unfamiliar with procedures to culture and study cells with these systems, with a particular focus on commercially available hydrogels.
Collapse
Affiliation(s)
- Steven R Caliari
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
462
|
Chang FC, Tsao CT, Lin A, Zhang M, Levengood SL, Zhang M. PEG-chitosan hydrogel with tunable stiffness for study of drug response of breast cancer cells. Polymers (Basel) 2016; 8:112. [PMID: 27595012 PMCID: PMC5004991 DOI: 10.3390/polym8040112] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 03/21/2016] [Indexed: 01/23/2023] Open
Abstract
Mechanical properties of the extracellular matrix have a profound effect on the behavior of anchorage-dependent cells. However, the mechanisms that define the effects of matrix stiffness on cell behavior remains unclear. Therefore, the development and fabrication of synthetic matrices with well-defined stiffness is invaluable for studying the interactions of cells with their biophysical microenvironment in vitro. We demonstrate a methoxypolyethylene glycol (mPEG)-modified chitosan hydrogel network where hydrogel stiffness can be easily modulated under physiological conditions by adjusting the degree of mPEG grafting onto chitosan (PEGylation). We show that the storage modulus of the hydrogel increases as PEGylation decreases and the gels exhibit instant self-recovery after deformation. Breast cancer cells cultured on the stiffest hydrogels adopt a more malignant phenotype with increased resistance to doxorubicin as compared with cells cultured on tissue culture polystyrene or Matrigel. This work demonstrates the utility of mPEG-modified chitosan hydrogel, with tunable mechanical properties, as an improved replacement of conventional culture system for in vitro characterization of breast cancer cell phenotype and evaluation of cancer therapies.
Collapse
Affiliation(s)
- Fei-Chien Chang
- Department of Materials Science and Engineering, University of Washington, 302L Roberts Hall, Seattle, WA 98195, USA; (F.-C.C.); (C.-T.T.); (A.L.); (S.L.L.)
| | - Ching-Ting Tsao
- Department of Materials Science and Engineering, University of Washington, 302L Roberts Hall, Seattle, WA 98195, USA; (F.-C.C.); (C.-T.T.); (A.L.); (S.L.L.)
| | - Anqi Lin
- Department of Materials Science and Engineering, University of Washington, 302L Roberts Hall, Seattle, WA 98195, USA; (F.-C.C.); (C.-T.T.); (A.L.); (S.L.L.)
| | - Mengying Zhang
- Department of Molecular Engineering and Science Institute, University of Washington, Seattle, WA 98195, USA;
| | - Sheeny Lan Levengood
- Department of Materials Science and Engineering, University of Washington, 302L Roberts Hall, Seattle, WA 98195, USA; (F.-C.C.); (C.-T.T.); (A.L.); (S.L.L.)
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, 302L Roberts Hall, Seattle, WA 98195, USA; (F.-C.C.); (C.-T.T.); (A.L.); (S.L.L.)
| |
Collapse
|
463
|
Sero JE, Sailem HZ, Ardy RC, Almuttaqi H, Zhang T, Bakal C. Cell shape and the microenvironment regulate nuclear translocation of NF-κB in breast epithelial and tumor cells. Mol Syst Biol 2016; 11:790. [PMID: 26148352 PMCID: PMC4380925 DOI: 10.15252/msb.20145644] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Although a great deal is known about the signaling events that promote nuclear translocation of NF-κB, how cellular biophysics and the microenvironment might regulate the dynamics of this pathway is poorly understood. In this study, we used high-content image analysis and Bayesian network modeling to ask whether cell shape and context features influence NF-κB activation using the inherent variability present in unperturbed populations of breast tumor and non-tumor cell lines. Cell–cell contact, cell and nuclear area, and protrusiveness all contributed to variability in NF-κB localization in the absence and presence of TNFα. Higher levels of nuclear NF-κB were associated with mesenchymal-like versus epithelial-like morphologies, and RhoA-ROCK-myosin II signaling was critical for mediating shape-based differences in NF-κB localization and oscillations. Thus, mechanical factors such as cell shape and the microenvironment can influence NF-κB signaling and may in part explain how different phenotypic outcomes can arise from the same chemical cues.
Collapse
Affiliation(s)
- Julia E Sero
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer ResearchLondon, UK
- * Corresponding author. Tel: +44 207 153 5170; E-mail:
| | - Heba Zuhair Sailem
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer ResearchLondon, UK
| | - Rico Chandra Ardy
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer ResearchLondon, UK
| | - Hannah Almuttaqi
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer ResearchLondon, UK
| | - Tongli Zhang
- Oxford Centre for Integrative Systems Biology, Department of Biochemistry, University of OxfordOxford, UK
| | - Chris Bakal
- Chester Beatty Laboratories, Division of Cancer Biology, Institute of Cancer ResearchLondon, UK
- ** Corresponding author. Tel: +44 207 153 5080; E-mail:
| |
Collapse
|
464
|
Muhamed I, Wu J, Sehgal P, Kong X, Tajik A, Wang N, Leckband DE. E-cadherin-mediated force transduction signals regulate global cell mechanics. J Cell Sci 2016; 129:1843-54. [PMID: 26966187 DOI: 10.1242/jcs.185447] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 03/03/2016] [Indexed: 12/22/2022] Open
Abstract
This report elucidates an E-cadherin-based force-transduction pathway that triggers changes in cell mechanics through a mechanism requiring epidermal growth factor receptor (EGFR), phosphoinositide 3-kinase (PI3K), and the downstream formation of new integrin adhesions. This mechanism operates in addition to local cytoskeletal remodeling triggered by conformational changes in the E-cadherin-associated protein α-catenin, at sites of mechanical perturbation. Studies using magnetic twisting cytometry (MTC), together with traction force microscopy (TFM) and confocal imaging identified force-activated E-cadherin-specific signals that integrate cadherin force transduction, integrin activation and cell contractility. EGFR is required for the downstream activation of PI3K and myosin-II-dependent cell stiffening. Our findings also demonstrated that α-catenin-dependent cytoskeletal remodeling at perturbed E-cadherin adhesions does not require cell stiffening. These results broaden the repertoire of E-cadherin-based force transduction mechanisms, and define the force-sensitive signaling network underlying the mechano-chemical integration of spatially segregated adhesion receptors.
Collapse
Affiliation(s)
- Ismaeel Muhamed
- Department of Biochemistry, University of Illinois Urbana Champaign, Urbana, IL 61801, USA
| | - Jun Wu
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana Champaign, Urbana, IL 61801, USA
| | - Poonam Sehgal
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana Champaign, Urbana, IL 61801, USA
| | - Xinyu Kong
- Department of Biochemistry, University of Illinois Urbana Champaign, Urbana, IL 61801, USA
| | - Arash Tajik
- Department of Mechanical Science and Engineering, University of Illinois Urbana Champaign, Urbana, IL 61801, USA
| | - Ning Wang
- Department of Mechanical Science and Engineering, University of Illinois Urbana Champaign, Urbana, IL 61801, USA
| | - Deborah E Leckband
- Department of Biochemistry, University of Illinois Urbana Champaign, Urbana, IL 61801, USA Department of Chemical and Biomolecular Engineering, University of Illinois Urbana Champaign, Urbana, IL 61801, USA Department of Chemistry, University of Illinois Urbana Champaign, Urbana, IL 61801, USA Carl W. Woese Institute of Genomic Biology, University of Illinois Urbana Champaign, Urbana, IL 61801, USA
| |
Collapse
|
465
|
Szeto SG, Narimatsu M, Lu M, He X, Sidiqi AM, Tolosa MF, Chan L, De Freitas K, Bialik JF, Majumder S, Boo S, Hinz B, Dan Q, Advani A, John R, Wrana JL, Kapus A, Yuen DA. YAP/TAZ Are Mechanoregulators of TGF- β-Smad Signaling and Renal Fibrogenesis. J Am Soc Nephrol 2016; 27:3117-3128. [PMID: 26961347 DOI: 10.1681/asn.2015050499] [Citation(s) in RCA: 334] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 02/02/2016] [Indexed: 11/03/2022] Open
Abstract
Like many organs, the kidney stiffens after injury, a process that is increasingly recognized as an important driver of fibrogenesis. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are related mechanosensory proteins that bind to Smad transcription factors, the canonical mediators of profibrotic TGF-β responses. Here, we investigated the role of YAP/TAZ in the matrix stiffness dependence of fibroblast responses to TGF-β In contrast to growth on a stiff surface, fibroblast growth on a soft matrix led to YAP/TAZ sequestration in the cytosol and impaired TGF-β-induced Smad2/3 nuclear accumulation and transcriptional activity. YAP knockdown or treatment with verteporfin, a drug that was recently identified as a potent YAP inhibitor, elicited similar changes. Furthermore, verteporfin reduced YAP/TAZ levels and decreased the total cellular levels of Smad2/3 after TGF-β stimulation. Verteporfin treatment of mice subjected to unilateral ureteral obstruction similarly reduced YAP/TAZ levels and nuclear Smad accumulation in the kidney, and attenuated renal fibrosis. Our data suggest that organ stiffening cooperates with TGF-β to induce fibrosis in a YAP/TAZ- and Smad2/3-dependent manner. Interference with this YAP/TAZ and TGF-β/Smad crosstalk likely underlies the antifibrotic activity of verteporfin. Finally, through repurposing of a clinically used drug, we illustrate the therapeutic potential of a novel mechanointerference strategy that blocks TGF-β signaling and renal fibrogenesis.
Collapse
Affiliation(s)
- Stephen G Szeto
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and.,Institute of Medical Science and
| | - Masahiro Narimatsu
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics
| | - Mingliang Lu
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and
| | - Xiaolin He
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and
| | - Ahmad M Sidiqi
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and.,Institute of Medical Science and
| | - Monica F Tolosa
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and.,Department of Laboratory Medicine and Pathobiology, School of Graduate Studies
| | - Lauren Chan
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and
| | - Krystale De Freitas
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and
| | - Janne Folke Bialik
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and
| | - Syamantak Majumder
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and
| | - Stellar Boo
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, and
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, and
| | - Qinghong Dan
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and.,Institute of Medical Science and
| | - Rohan John
- Department of Laboratory Medicine and Pathobiology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey L Wrana
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics
| | - Andras Kapus
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and.,Institute of Medical Science and
| | - Darren A Yuen
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; and .,Institute of Medical Science and.,Department of Laboratory Medicine and Pathobiology, School of Graduate Studies
| |
Collapse
|
466
|
Cell shape dynamics reveal balance of elasticity and contractility in peripheral arcs. Biophys J 2016; 108:2437-2447. [PMID: 25992722 DOI: 10.1016/j.bpj.2015.04.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 02/11/2015] [Accepted: 04/01/2015] [Indexed: 01/04/2023] Open
Abstract
The mechanical interaction between adherent cells and their substrate relies on the formation of adhesion sites and on the stabilization of contractile acto-myosin bundles, or stress fibers. The shape of the cell and the orientation of these fibers can be controlled by adhesive patterning. On nonadhesive gaps, fibroblasts develop thick peripheral stress fibers, with a concave curvature. The radius of curvature of these arcs results from the balance of the line tension in the arc and of the surface tension in the cell bulk. However, the nature of these forces, and in particular the contribution of myosin-dependent contractility, is not clear. To get insight into the force balance, we inhibit myosin activity and simultaneously monitor the dynamics of peripheral arc radii and traction forces. We use these measurements to estimate line and surface tension. We found that myosin inhibition led to a decrease in the traction forces and an increase in arc radius, indicating that both line tension and surface tension dropped, but the line tension decreased to a lesser extent than surface tension. These results suggest that myosin-independent force contributes to tension in the peripheral arcs. We propose a simple physical model in which the peripheral arc line tension is due to the combination of myosin II contractility and a passive elastic component, while surface tension is largely due to active contractility. Numerical solutions of this model reproduce well the experimental data and allow estimation of the contributions of elasticity and contractility to the arc line tension.
Collapse
|
467
|
Uto K, Mano SS, Aoyagi T, Ebara M. Substrate Fluidity Regulates Cell Adhesion and Morphology on Poly(ε-caprolactone)-Based Materials. ACS Biomater Sci Eng 2016; 2:446-453. [DOI: 10.1021/acsbiomaterials.6b00058] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Koichiro Uto
- Biomaterials
Unit, International
Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Sharmy S. Mano
- Biomaterials
Unit, International
Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Takao Aoyagi
- Biomaterials
Unit, International
Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Mitsuhiro Ebara
- Biomaterials
Unit, International
Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| |
Collapse
|
468
|
Chevalier N, Gazguez E, Bidault L, Guilbert T, Vias C, Vian E, Watanabe Y, Muller L, Germain S, Bondurand N, Dufour S, Fleury V. How Tissue Mechanical Properties Affect Enteric Neural Crest Cell Migration. Sci Rep 2016; 6:20927. [PMID: 26887292 PMCID: PMC4757826 DOI: 10.1038/srep20927] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 01/13/2016] [Indexed: 12/19/2022] Open
Abstract
Neural crest cells (NCCs) are a population of multipotent cells that migrate extensively during vertebrate development. Alterations to neural crest ontogenesis cause several diseases, including cancers and congenital defects, such as Hirschprung disease, which results from incomplete colonization of the colon by enteric NCCs (ENCCs). We investigated the influence of the stiffness and structure of the environment on ENCC migration in vitro and during colonization of the gastrointestinal tract in chicken and mouse embryos. We showed using tensile stretching and atomic force microscopy (AFM) that the mesenchyme of the gut was initially soft but gradually stiffened during the period of ENCC colonization. Second-harmonic generation (SHG) microscopy revealed that this stiffening was associated with a gradual organization and enrichment of collagen fibers in the developing gut. Ex-vivo 2D cell migration assays showed that ENCCs migrated on substrates with very low levels of stiffness. In 3D collagen gels, the speed of the ENCC migratory front decreased with increasing gel stiffness, whereas no correlation was found between porosity and ENCC migration behavior. Metalloprotease inhibition experiments showed that ENCCs actively degraded collagen in order to progress. These results shed light on the role of the mechanical properties of tissues in ENCC migration during development.
Collapse
Affiliation(s)
- N.R. Chevalier
- Laboratoire Matière et Systèmes Complexes, Université Paris-Diderot/CNRS UMR 7057, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| | - E. Gazguez
- UMR144, CNRS-Institut Curie, 26, rue d’Ulm, 75248 Paris cedex 05, France
| | - L. Bidault
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris, F-75005, France
- INSERM, U1050, Paris, F-75005, France
- CNRS, UMR 7241, Paris, F-75005, France
| | - T. Guilbert
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - C. Vias
- Laboratoire Matière et Systèmes Complexes, Université Paris-Diderot/CNRS UMR 7057, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| | - E. Vian
- Laboratoire Matière et Systèmes Complexes, Université Paris-Diderot/CNRS UMR 7057, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| | - Y. Watanabe
- INSERM U955, Equipe 11, F-94000 Créteil, France
| | - L. Muller
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris, F-75005, France
- INSERM, U1050, Paris, F-75005, France
- CNRS, UMR 7241, Paris, F-75005, France
| | - S. Germain
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB), Paris, F-75005, France
- INSERM, U1050, Paris, F-75005, France
- CNRS, UMR 7241, Paris, F-75005, France
| | | | - S. Dufour
- UMR144, CNRS-Institut Curie, 26, rue d’Ulm, 75248 Paris cedex 05, France
| | - V. Fleury
- Laboratoire Matière et Systèmes Complexes, Université Paris-Diderot/CNRS UMR 7057, 10 rue Alice Domon et Léonie Duquet, 75013 Paris, France
| |
Collapse
|
469
|
Lourenço T, Paes de Faria J, Bippes CA, Maia J, Lopes-da-Silva JA, Relvas JB, Grãos M. Modulation of oligodendrocyte differentiation and maturation by combined biochemical and mechanical cues. Sci Rep 2016; 6:21563. [PMID: 26879561 PMCID: PMC4754901 DOI: 10.1038/srep21563] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 01/25/2016] [Indexed: 01/17/2023] Open
Abstract
Extracellular matrix (ECM) proteins play a key role during oligodendrogenesis. While fibronectin (FN) is involved in the maintenance and proliferation of oligodendrocyte progenitor cells (OPCs), merosin (MN) promotes differentiation into oligodendrocytes (OLs). Mechanical properties of the ECM also seem to affect OL differentiation, hence this study aimed to clarify the impact of combined biophysical and biochemical elements during oligodendrocyte differentiation and maturation using synthetic elastic polymeric ECM-like substrates. CG-4 cells presented OPC- or OL-like morphology in response to brain-compliant substrates functionalised with FN or MN, respectively. The expression of the differentiation and maturation markers myelin basic protein — MBP — and proteolipid protein — PLP — (respectively) by primary rat oligodendrocytes was enhanced in presence of MN, but only on brain-compliant conditions, considering the distribution (MBP) or amount (PLP) of the protein. It was also observed that maturation of OLs was attained earlier (by assessing PLP expression) by cells differentiated on MN-functionalised brain-compliant substrates than on standard culture conditions. Moreover, the combination of MN and substrate compliance enhanced the maturation and morphological complexity of OLs. Considering the distinct degrees of stiffness tested ranging within those of the central nervous system, our results indicate that 6.5 kPa is the most suitable rigidity for oligodendrocyte differentiation.
Collapse
Affiliation(s)
- Tânia Lourenço
- Biocant, Technology Transfer Association, Cantanhede, Portugal.,Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Joana Paes de Faria
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | | | - João Maia
- Chemical Engineering Department, Faculty of Science and Technology, University of Coimbra, Coimbra, Portugal
| | | | - João B Relvas
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - Mário Grãos
- Biocant, Technology Transfer Association, Cantanhede, Portugal.,Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
470
|
Ko P, Kim D, You E, Jung J, Oh S, Kim J, Lee KH, Rhee S. Extracellular Matrix Rigidity-dependent Sphingosine-1-phosphate Secretion Regulates Metastatic Cancer Cell Invasion and Adhesion. Sci Rep 2016; 6:21564. [PMID: 26877098 PMCID: PMC4753492 DOI: 10.1038/srep21564] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/27/2016] [Indexed: 12/14/2022] Open
Abstract
Dynamic interaction between cancer cells and the surrounding microenvironment is critical for cancer progression via changes in cellular behavior including alteration of secreted molecules. However, the molecular mechanisms underlying the influence exerted by the cancer microenvironment on secretion of molecules during cancer progression remain largely unknown. In this study, we report that secretion of spingsine-1-phosphate (S1P) and its regulator, SphK1 expression is dependent of the substrate rigidity, which is critical for the balance between cancer cell invasion and adhesion. Conditioned media (CM) of MDA-MB-231, an aggressive breast cancer cell obtained from soft substrate (~0.5 kPa) induced chemo-attractive invasion, while CM obtained from stiff substrate (~2.5 kPa) increased cell adhesion instead. We found that the expression of SphK1 is upregulated in the stiff substrate, resulting in an increase in S1P levels in the CM. We also found that upregulation of SphK1 expression in the stiff substrate is dominant in metastatic cancer cells but not in primary cancer cells. These results suggest that alterations in the mechanical environment of the ECM surrounding the tumor cells actively regulate cellular properties such as secretion, which in turn, may contribute to cancer progression.
Collapse
Affiliation(s)
- Panseon Ko
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Daehwan Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Eunae You
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jangho Jung
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Somi Oh
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jaehyun Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Kwang-Ho Lee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| |
Collapse
|
471
|
Rao SR, Subbarayan R, Dinesh MG, Arumugam G, Raja STK. Differentiation of human gingival mesenchymal stem cells into neuronal lineages in 3D bioconjugated injectable protein hydrogel construct for the management of neuronal disorder. Exp Mol Med 2016; 48:e209. [PMID: 26869025 PMCID: PMC4892868 DOI: 10.1038/emm.2015.113] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/14/2015] [Accepted: 09/30/2015] [Indexed: 01/05/2023] Open
Abstract
The success of regeneration attempt is based on an ideal combination of stem cells, scaffolding and growth factors. Tissue constructs help to maintain stem cells in a required area for a desired time. There is a need for easily obtainable cells, potentially autologous stem cells and a biologically acceptable scaffold for use in humans in different difficult situations. This study aims to address these issues utilizing a unique combination of stem cells from gingiva and a hydrogel scaffold, based on a natural product for regenerative application. Human gingival mesenchymal stem cells (HGMSCs) were, with due induction, differentiated to neuronal lineages to overcome the problems associated with birth tissue-related stem cells. The differentiation potential of neuronal lineages was confirmed with suitable specific markers. The properties of mesenchymal stem cells in encapsulated form were observed to be similar to free cells. The encapsulated cells (3D) were then subjected to differentiation into neuronal lineages with suitable inducers, and the morphology and gene expression of transient cells were analyzed. HGMSCs was differentiated into neuronal lineages as both free and encapsulated forms without any significant differences. The presence of Nissl bodies and the neurite outgrowth confirm the differentiation. The advantages of this new combination appear to make it a promising tissue construct for translational application.
Collapse
Affiliation(s)
- Suresh Ranga Rao
- Department of Periodontology and Implantology, Faculty of Dental Sciences, Centre for Regenerative Medicine and Stem Cell Research, Sri Ramachandra University, Chennai, India
| | - Rajasekaran Subbarayan
- Centre for Regenerative Medicine and Stem Cell Research, Central Research Facility, Sri Ramachandra University, Chennai, India
| | - Murugan Girija Dinesh
- Centres for Indian Systems of Medicine Quality Assurance and Standardization, Sri Ramachandra University, Chennai, India
| | - Gnanamani Arumugam
- Microbiology Division, Central Leather Research Institute Adyar, Chennai, India
| | | |
Collapse
|
472
|
Ribeiro AJS, Denisin AK, Wilson RE, Pruitt BL. For whom the cells pull: Hydrogel and micropost devices for measuring traction forces. Methods 2016; 94:51-64. [PMID: 26265073 PMCID: PMC4746112 DOI: 10.1016/j.ymeth.2015.08.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 07/10/2015] [Accepted: 08/06/2015] [Indexed: 01/16/2023] Open
Abstract
While performing several functions, adherent cells deform their surrounding substrate via stable adhesions that connect the intracellular cytoskeleton to the extracellular matrix. The traction forces that deform the substrate are studied in mechanotrasduction because they are affected by the mechanics of the extracellular milieu. We review the development and application of two methods widely used to measure traction forces generated by cells on 2D substrates: (i) traction force microscopy with polyacrylamide hydrogels and (ii) calculation of traction forces with arrays of deformable microposts. Measuring forces with these methods relies on measuring substrate displacements and converting them into forces. We describe approaches to determine force from displacements and elaborate on the necessary experimental conditions for this type of analysis. We emphasize device fabrication, mechanical calibration of substrates and covalent attachment of extracellular matrix proteins to substrates as key features in the design of experiments to measure cell traction forces with polyacrylamide hydrogels or microposts. We also report the challenges and achievements in integrating these methods with platforms for the mechanical stimulation of adherent cells. The approaches described here will enable new studies to understand cell mechanical outputs as a function of mechanical inputs and advance the understanding of mechanotransduction mechanisms.
Collapse
Affiliation(s)
- Alexandre J S Ribeiro
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States; Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, United States
| | - Aleksandra K Denisin
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States; Stanford Bioengineering, Stanford University, Stanford, CA 94305, United States
| | - Robin E Wilson
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States
| | - Beth L Pruitt
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States; Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, United States; Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA 94305, United States.
| |
Collapse
|
473
|
|
474
|
Stoppel WL, Kaplan DL, Black LD. Electrical and mechanical stimulation of cardiac cells and tissue constructs. Adv Drug Deliv Rev 2016; 96:135-55. [PMID: 26232525 DOI: 10.1016/j.addr.2015.07.009] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/16/2015] [Accepted: 07/25/2015] [Indexed: 12/19/2022]
Abstract
The field of cardiac tissue engineering has made significant strides over the last few decades, highlighted by the development of human cell derived constructs that have shown increasing functional maturity over time, particularly using bioreactor systems to stimulate the constructs. However, the functionality of these tissues is still unable to match that of native cardiac tissue and many of the stem-cell derived cardiomyocytes display an immature, fetal like phenotype. In this review, we seek to elucidate the biological underpinnings of both mechanical and electrical signaling, as identified via studies related to cardiac development and those related to an evaluation of cardiac disease progression. Next, we review the different types of bioreactors developed to individually deliver electrical and mechanical stimulation to cardiomyocytes in vitro in both two and three-dimensional tissue platforms. Reactors and culture conditions that promote functional cardiomyogenesis in vitro are also highlighted. We then cover the more recent work in the development of bioreactors that combine electrical and mechanical stimulation in order to mimic the complex signaling environment present in vivo. We conclude by offering our impressions on the important next steps for physiologically relevant mechanical and electrical stimulation of cardiac cells and engineered tissue in vitro.
Collapse
|
475
|
Lesman A, Rosenfeld D, Landau S, Levenberg S. Mechanical regulation of vascular network formation in engineered matrices. Adv Drug Deliv Rev 2016. [PMID: 26212159 DOI: 10.1016/j.addr.2015.07.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Generation of vessel networks within engineered tissues is critical for integration and perfusion of the implanted tissue in vivo. The effect of mechanical cues in guiding and stabilizing the vessels has begun to attract marked interest. This review surveys the impact of mechanical cues on formation of vascular networks in 2D and 3D gel matrices. We give less emphasis to regulation of endothelial monolayers and single endothelial cells. Several vascularization models have consistently found that the stress generated in the gel, and encountered by embedded cells, control various aspects of vascular network formation, including sprouting, branching, alignment, and vessel maturation. This internal stress is generated by cell contractile forces, and is balanced by gel stiffness and boundary constrains imposed on the gel. Actin and myosin II are key molecular players in controlling initiation of vessel sprouting and branching morphogenesis. Additionally, the impact of external mechanical cues on tissue vascularization, and studies supporting the notion that mechanical forces regulate vascularization in the live animal are reviewed.
Collapse
Affiliation(s)
- Ayelet Lesman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel; School of Mechanical Engineering, Faculty of Engineering, Tel-Aviv University, Israel
| | - Dekel Rosenfeld
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Israel
| | - Shira Landau
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Israel
| | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Israel.
| |
Collapse
|
476
|
Varney SD, Betts CB, Zheng R, Wu L, Hinz B, Zhou J, Van De Water L. Hic-5 is required for myofibroblast differentiation by regulating mechanically dependent MRTF-A nuclear accumulation. J Cell Sci 2016; 129:774-87. [PMID: 26759173 DOI: 10.1242/jcs.170589] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 01/04/2016] [Indexed: 01/21/2023] Open
Abstract
How mechanical cues from the extracellular environment are translated biochemically to modulate the effects of TGF-β on myofibroblast differentiation remains a crucial area of investigation. We report here that the focal adhesion protein, Hic-5 (also known as TGFB1I1), is required for the mechanically dependent generation of stress fibers in response to TGF-β. Successful generation of stress fibers promotes the nuclear localization of the transcriptional co-factor MRTF-A (also known as MKL1), and this correlates with the mechanically dependent induction of α smooth muscle actin (α-SMA) and Hic-5 in response to TGF-β. As a consequence of regulating stress fiber assembly, Hic-5 is required for the nuclear accumulation of MRTF-A and the induction of α-SMA as well as cellular contractility, suggesting a crucial role for Hic-5 in myofibroblast differentiation. Indeed, the expression of Hic-5 was transient in acute wounds and persistent in pathogenic scars, and Hic-5 colocalized with α-SMA expression in vivo. Taken together, these data suggest that a mechanically dependent feed-forward loop, elaborated by the reciprocal regulation of MRTF-A localization by Hic-5 and Hic-5 expression by MRTF-A, plays a crucial role in myofibroblast differentiation in response to TGF-β.
Collapse
Affiliation(s)
- Scott D Varney
- Center for Cell Biology and Cancer Research (MC-165), Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA
| | - Courtney B Betts
- Center for Cell Biology and Cancer Research (MC-165), Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA
| | - Rui Zheng
- Center for Cell Biology and Cancer Research (MC-165), Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA
| | - Lei Wu
- Center for Cell Biology and Cancer Research (MC-165), Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, 150 College Street, FitzGerald Building, Room 234, Toronto, Ontario, Canada M5S 3E2
| | - Jiliang Zhou
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, CB-3628, 1459 Laney Walker Boulevard, Augusta, GA 30912, USA
| | - Livingston Van De Water
- Center for Cell Biology and Cancer Research (MC-165), Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA
| |
Collapse
|
477
|
Modulating the Substrate Stiffness to Manipulate Differentiation of Resident Liver Stem Cells and to Improve the Differentiation State of Hepatocytes. Stem Cells Int 2016; 2016:5481493. [PMID: 27057172 PMCID: PMC4737459 DOI: 10.1155/2016/5481493] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 12/14/2022] Open
Abstract
In many cell types, several cellular processes, such as differentiation of stem/precursor cells, maintenance of differentiated phenotype, motility, adhesion, growth, and survival, strictly depend on the stiffness of extracellular matrix that, in vivo, characterizes their correspondent organ and tissue. In the liver, the stromal rigidity is essential to obtain the correct organ physiology whereas any alteration causes liver cell dysfunctions. The rigidity of the substrate is an element no longer negligible for the cultivation of several cell types, so that many data so far obtained, where cells have been cultured on plastic, could be revised. Regarding liver cells, standard culture conditions lead to the dedifferentiation of primary hepatocytes, transdifferentiation of stellate cells into myofibroblasts, and loss of fenestration of sinusoidal endothelium. Furthermore, standard cultivation of liver stem/precursor cells impedes an efficient execution of the epithelial/hepatocyte differentiation program, leading to the expansion of a cell population expressing only partially liver functions and products. Overcoming these limitations is mandatory for any approach of liver tissue engineering. Here we propose cell lines as in vitro models of liver stem cells and hepatocytes and an innovative culture method that takes into account the substrate stiffness to obtain, respectively, a rapid and efficient differentiation process and the maintenance of the fully differentiated phenotype.
Collapse
|
478
|
Minaisah RM, Cox S, Warren DT. The Use of Polyacrylamide Hydrogels to Study the Effects of Matrix Stiffness on Nuclear Envelope Properties. Methods Mol Biol 2016; 1411:233-9. [PMID: 27147046 DOI: 10.1007/978-1-4939-3530-7_15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Matrix-derived mechanical cues influence cell proliferation, motility, and differentiation. Recent findings clearly demonstrate that the nuclear envelope (NE) adapts and remodels in response to mechanical signals, including matrix stiffness, yet a plethora of studies have been performed on tissue culture plastic or glass that have a similar stiffness to cortical bone. Using methods that allow modulation of matrix stiffness will provide further insight into the role of the NE in physiological conditions and the impact of changes in stiffness observed during ageing and disease on cellular function. In this chapter, we describe the polyacrylamide hydrogel system, which allows fabrication of hydrogels with variable stiffness to better mimic the environment experienced by cells in most tissues of the body.
Collapse
Affiliation(s)
- Rose-Marie Minaisah
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, James Black Centre, King's College London, London, SE5 9NU, UK
| | - Susan Cox
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, King's College London, London, SE1 1UL, UK
| | - Derek T Warren
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, James Black Centre, King's College London, London, SE5 9NU, UK.
| |
Collapse
|
479
|
Contractile dynamics change before morphological cues during fluorescence [corrected] illumination. Sci Rep 2015; 5:18513. [PMID: 26691776 PMCID: PMC4686977 DOI: 10.1038/srep18513] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/19/2015] [Indexed: 01/15/2023] Open
Abstract
Illumination can have adverse effects on live cells. However, many experiments, e.g. traction force microscopy, rely on fluorescence microscopy. Current methods to assess undesired photo-induced cell changes rely on qualitative observation of changes in cell morphology. Here we utilize a quantitative technique to identify the effect of light on cell contractility prior to morphological changes. Fibroblasts were cultured on soft elastic hydrogels embedded with fluorescent beads. The adherent cells generated contractile forces that deform the substrate. Beads were used as fiducial markers to quantify the substrate deformation over time, which serves as a measure of cell force dynamics. We find that cells exposed to moderate fluorescence illumination (λ = 540–585 nm, I = 12.5 W/m2, duration = 60 s) exhibit rapid force relaxation. Strikingly, cells exhibit force relaxation after only 2 s of exposure, suggesting that photo-induced relaxation occurs nearly immediately. Evidence of photo-induced morphological changes were not observed for 15–30 min after illumination. Force relaxation and morphological changes were found to depend on wavelength and intensity of excitation light. This study demonstrates that changes in cell contractility reveal evidence of a photo-induced cell response long before any morphological cues.
Collapse
|
480
|
Acevedo-Acevedo S, Crone WC. Substrate stiffness effect and chromosome missegregation in hIPS cells. J Negat Results Biomed 2015; 14:22. [PMID: 26683848 PMCID: PMC4683860 DOI: 10.1186/s12952-015-0042-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 12/10/2015] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Ensuring genetic stability in pluripotent stem cell (PSC) cultures is essential for the development of successful cell therapies. Although most instances lead to failed experiments and go unreported in the literature, many laboratories have found the emergence of genetic abnormalities in PSCs when cultured in vitro for prolonged amounts of time. These cells are primarily cultured in non-physiological stiff substrates like tissue culture polystyrene (TCPS) which raises the possibility that the cause of these abnormalities may be influenced by substrate mechanics. FINDINGS In order to investigate this, human PSCs were grown on substrates of varying stiffness such as a range of polyacrylamide formulations, TCPS, and borosilicate glass coverslips. These substrates allowed for the testing of a stiffness range from 5kPa to 64GPa. Two human induced PSC (iPSC) lines were analyzed in this study: 19-9-11 iPSCs and 19.7 clone F iPSCs. Centrosome and DNA staining revealed that 19-9-11 iPSCs range from 1-8.5 % abnormal mitoses under the different culture conditions. A range of 4.4-8.1 % abnormal mitoses was found for 19.7 clone F iPSCs. CONCLUSIONS Abnormal cell division was not biased towards one particular substrate. It was confirmed by Analysis of Variance (ANOVA) and Tukey's Honest Significant Difference test that there was no statistically significant difference between passage numbers, cell lines, or substrates.
Collapse
Affiliation(s)
| | - Wendy C Crone
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA. .,Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
481
|
Visualization of adherent cell monolayers by cryo-electron microscopy: A snapshot of endothelial adherens junctions. J Struct Biol 2015; 192:470-477. [DOI: 10.1016/j.jsb.2015.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 01/05/2023]
|
482
|
Ilin Y, Choi JS, Harley BAC, Kraft ML. Identifying States along the Hematopoietic Stem Cell Differentiation Hierarchy with Single Cell Specificity via Raman Spectroscopy. Anal Chem 2015; 87:11317-24. [PMID: 26496164 PMCID: PMC4687963 DOI: 10.1021/acs.analchem.5b02537] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A major challenge for expanding specific types of hematopoietic cells ex vivo for the treatment of blood cell pathologies is identifying the combinations of cellular and matrix cues that direct hematopoietic stem cells (HSC) to self-renew or differentiate into cell populations ex vivo. Microscale screening platforms enable minimizing the number of rare HSCs required to screen the effects of numerous cues on HSC fate decisions. These platforms create a strong demand for label-free methods that accurately identify the fate decisions of individual hematopoietic cells at specific locations on the platform. We demonstrate the capacity to identify discrete cells along the HSC differentiation hierarchy via multivariate analysis of Raman spectra. Notably, cell state identification is accurate for individual cells and independent of the biophysical properties of the functionalized polyacrylamide gels upon which these cells are cultured. We report partial least-squares discriminant analysis (PLS-DA) models of single cell Raman spectra enable identifying four dissimilar hematopoietic cell populations across the HSC lineage specification. Successful discrimination was obtained for a population enriched for long-term repopulating HSCs (LT-HSCs) versus their more differentiated progeny, including closely related short-term repopulating HSCs (ST-HSCs) and fully differentiated lymphoid (B cells) and myeloid (granulocytes) cells. The lineage-specific differentiation states of cells from these four subpopulations were accurately identified independent of the stiffness of the underlying biomaterial substrate, indicating subtle spectral variations that discriminated these populations were not masked by features from the culture substrate. This approach enables identifying the lineage-specific differentiation stages of hematopoietic cells on biomaterial substrates of differing composition and may facilitate correlating hematopoietic cell fate decisions with the extrinsic cues that elicited them.
Collapse
Affiliation(s)
- Yelena Ilin
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Ji Sun Choi
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Brendan A. C. Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Carle R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Mary L. Kraft
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
483
|
Álvarez-González B, Meili R, Bastounis E, Firtel RA, Lasheras JC, Del Álamo JC. Three-dimensional balance of cortical tension and axial contractility enables fast amoeboid migration. Biophys J 2015; 108:821-832. [PMID: 25692587 DOI: 10.1016/j.bpj.2014.11.3478] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 11/20/2014] [Accepted: 11/21/2014] [Indexed: 11/25/2022] Open
Abstract
Fast amoeboid migration requires cells to apply mechanical forces on their surroundings via transient adhesions. However, the role these forces play in controlling cell migration speed remains largely unknown. We used three-dimensional force microscopy to measure the three-dimensional forces exerted by chemotaxing Dictyostelium cells, and examined wild-type cells as well as mutants with defects in contractility, internal F-actin crosslinking, and cortical integrity. We showed that cells pull on their substrate adhesions using two distinct, yet interconnected mechanisms: axial actomyosin contractility and cortical tension. We found that the migration speed increases when axial contractility overcomes cortical tension to produce the cell shape changes needed for locomotion. We demonstrated that the three-dimensional pulling forces generated by both mechanisms are internally balanced by an increase in cytoplasmic pressure that allows cells to push on their substrate without adhering to it, and which may be relevant for amoeboid migration in complex three-dimensional environments.
Collapse
Affiliation(s)
- Begoña Álvarez-González
- Department of Mechanical and Aerospace Engineering, University of California at San Diego, San Diego, California; Cell and Developmental Biology, Division of Biological Sciences, University of California at San Diego, San Diego, California
| | - Ruedi Meili
- Cell and Developmental Biology, Division of Biological Sciences, University of California at San Diego, San Diego, California
| | - Effie Bastounis
- Department of Mechanical and Aerospace Engineering, University of California at San Diego, San Diego, California; Cell and Developmental Biology, Division of Biological Sciences, University of California at San Diego, San Diego, California
| | - Richard A Firtel
- Cell and Developmental Biology, Division of Biological Sciences, University of California at San Diego, San Diego, California
| | - Juan C Lasheras
- Department of Mechanical and Aerospace Engineering, University of California at San Diego, San Diego, California; Department of Bioengineering, University of California at San Diego, San Diego, California; Institute for Engineering in Medicine, University of California at San Diego, San Diego, California
| | - Juan C Del Álamo
- Department of Mechanical and Aerospace Engineering, University of California at San Diego, San Diego, California; Institute for Engineering in Medicine, University of California at San Diego, San Diego, California.
| |
Collapse
|
484
|
Lin CH, Pelissier FA, Zhang H, Lakins J, Weaver VM, Park C, LaBarge MA. Microenvironment rigidity modulates responses to the HER2 receptor tyrosine kinase inhibitor lapatinib via YAP and TAZ transcription factors. Mol Biol Cell 2015; 26:3946-53. [PMID: 26337386 PMCID: PMC4710228 DOI: 10.1091/mbc.e15-07-0456] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 08/12/2015] [Accepted: 08/26/2015] [Indexed: 12/31/2022] Open
Abstract
Stiffness is a biophysical property of the extracellular matrix that modulates cellular functions, including proliferation, invasion, and differentiation, and it also may affect therapeutic responses. Therapeutic durability in cancer treatments remains a problem for both chemotherapies and pathway-targeted drugs, but the reasons for this are not well understood. Tumor progression is accompanied by changes in the biophysical properties of the tissue, and we asked whether matrix rigidity modulated the sensitive versus resistant states in HER2-amplified breast cancer cell responses to the HER2-targeted kinase inhibitor lapatinib. The antiproliferative effect of lapatinib was inversely proportional to the elastic modulus of the adhesive substrata. Down-regulation of the mechanosensitive transcription coactivators YAP and TAZ, either by siRNA or with the small-molecule YAP/TEAD inhibitor verteporfin, eliminated modulus-dependent lapatinib resistance. Reduction of YAP in vivo in mice also slowed the growth of implanted HER2-amplified tumors, showing a trend of increasing sensitivity to lapatinib as YAP decreased. Thus we address the role of stiffness in resistance to and efficacy of a HER2 pathway-targeted therapeutic via the mechanotransduction arm of the Hippo pathway.
Collapse
Affiliation(s)
- Chun-Han Lin
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 Program in Comparative Biochemistry, University of California, Berkeley, Berkeley, CA 94720
| | - Fanny A Pelissier
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720 Department of Biomedicine, University of Bergen, N-5009 Bergen, Norway
| | - Hui Zhang
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143
| | - Jon Lakins
- Center for Bioengineering, Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Valerie M Weaver
- Center for Bioengineering, Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Catherine Park
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143
| | - Mark A LaBarge
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| |
Collapse
|
485
|
Lee J, Abdeen AA, Tang X, Saif TA, Kilian KA. Geometric guidance of integrin mediated traction stress during stem cell differentiation. Biomaterials 2015; 69:174-83. [PMID: 26285084 PMCID: PMC4556610 DOI: 10.1016/j.biomaterials.2015.08.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 08/04/2015] [Indexed: 12/31/2022]
Abstract
Cells sense and transduce the chemical and mechanical properties of their microenvironment through cell surface integrin receptors. Traction stress exerted by cells on the extracellular matrix mediates focal adhesion stabilization and regulation of the cytoskeleton for directing biological activity. Understanding how stem cells integrate biomaterials properties through focal adhesions during differentiation is important for the design of soft materials for regenerative medicine. In this paper we use micropatterned hydrogels containing fluorescent beads to explore force transmission through integrins from single mesenchymal stem cells (MSCs) during differentiation. When cultured on polyacrylamide gels, MSCs will express markers associated with osteogenesis and myogenesis in a stiffness dependent manner. The shape of single cells and the composition of tethered matrix protein both influence the magnitude of traction stress applied and the resultant differentiation outcome. We show how geometry guides the spatial positioning of focal adhesions to maximize interaction with the matrix, and uncover a relationship between αvβ3, α5β1 and mechanochemical regulation of osteogenesis.
Collapse
Affiliation(s)
- Junmin Lee
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, IL 61801, USA; Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, IL 61801, USA
| | - Amr A Abdeen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, IL 61801, USA; Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, IL 61801, USA
| | - Xin Tang
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, IL 61801, USA; Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Taher A Saif
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, IL 61801, USA; Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kristopher A Kilian
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, IL 61801, USA; Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, IL 61801, USA.
| |
Collapse
|
486
|
Buffinton CM, Tong KJ, Blaho RA, Buffinton EM, Ebenstein DM. Comparison of mechanical testing methods for biomaterials: Pipette aspiration, nanoindentation, and macroscale testing. J Mech Behav Biomed Mater 2015; 51:367-79. [DOI: 10.1016/j.jmbbm.2015.07.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/17/2015] [Accepted: 07/22/2015] [Indexed: 10/23/2022]
|
487
|
Chiasson-MacKenzie C, Morris ZS, Baca Q, Morris B, Coker JK, Mirchev R, Jensen AE, Carey T, Stott SL, Golan DE, McClatchey AI. NF2/Merlin mediates contact-dependent inhibition of EGFR mobility and internalization via cortical actomyosin. J Cell Biol 2015; 211:391-405. [PMID: 26483553 PMCID: PMC4621825 DOI: 10.1083/jcb.201503081] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 09/01/2015] [Indexed: 01/04/2023] Open
Abstract
Merlin and Ezrin are central to a mechanism whereby mechanical forces transduced across the apical actomyosin cytoskeleton from cell junctions control the mobility and internalization of EGFR, providing novel insight into how cells inhibit mitogenic signaling in response to cell contact. The proliferation of normal cells is inhibited at confluence, but the molecular basis of this phenomenon, known as contact-dependent inhibition of proliferation, is unclear. We previously identified the neurofibromatosis type 2 (NF2) tumor suppressor Merlin as a critical mediator of contact-dependent inhibition of proliferation and specifically found that Merlin inhibits the internalization of, and signaling from, the epidermal growth factor receptor (EGFR) in response to cell contact. Merlin is closely related to the membrane–cytoskeleton linking proteins Ezrin, Radixin, and Moesin, and localization of Merlin to the cortical cytoskeleton is required for contact-dependent regulation of EGFR. We show that Merlin and Ezrin are essential components of a mechanism whereby mechanical forces associated with the establishment of cell–cell junctions are transduced across the cell cortex via the cortical actomyosin cytoskeleton to control the lateral mobility and activity of EGFR, providing novel insight into how cells inhibit mitogenic signaling in response to cell contact.
Collapse
Affiliation(s)
- Christine Chiasson-MacKenzie
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129 Department of Pathology, Massachusetts General Hospital, Charlestown, MA 02129 Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Zachary S Morris
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129 Department of Pathology, Massachusetts General Hospital, Charlestown, MA 02129 Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Quentin Baca
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115
| | - Brett Morris
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129 Department of Pathology, Massachusetts General Hospital, Charlestown, MA 02129 Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Joanna K Coker
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129 Department of Pathology, Massachusetts General Hospital, Charlestown, MA 02129 Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Rossen Mirchev
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115
| | - Anne E Jensen
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129 BioMEMs Resource Center, Massachusetts General Hospital, Charlestown, MA 02129
| | - Thomas Carey
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129 BioMEMs Resource Center, Massachusetts General Hospital, Charlestown, MA 02129
| | - Shannon L Stott
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129 BioMEMs Resource Center, Massachusetts General Hospital, Charlestown, MA 02129 Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - David E Golan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115
| | - Andrea I McClatchey
- Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA 02129 Department of Pathology, Massachusetts General Hospital, Charlestown, MA 02129 Department of Pathology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
488
|
The matrix protein Fibulin-5 is at the interface of tissue stiffness and inflammation in fibrosis. Nat Commun 2015; 6:8574. [PMID: 26469761 PMCID: PMC4634219 DOI: 10.1038/ncomms9574] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/04/2015] [Indexed: 12/11/2022] Open
Abstract
Fibrosis is a pervasive disease in which the excessive deposition of extracellular matrix (ECM) compromises tissue function. Although the underlying mechanisms are mostly unknown, matrix stiffness is increasingly appreciated as a contributor to fibrosis rather than merely a manifestation of the disease. Here we show that the loss of Fibulin-5, an elastic fibre component, not only decreases tissue stiffness, but also diminishes the inflammatory response and abrogates the fibrotic phenotype in a mouse model of cutaneous fibrosis. Increasing matrix stiffness raises the inflammatory response above a threshold level, independent of TGF-β, to stimulate further ECM secretion from fibroblasts and advance the progression of fibrosis. These results suggest that Fibulin-5 may be a therapeutic target to short-circuit this profibrotic feedback loop.
Collapse
|
489
|
Santos L, Fuhrmann G, Juenet M, Amdursky N, Horejs CM, Campagnolo P, Stevens MM. Extracellular Stiffness Modulates the Expression of Functional Proteins and Growth Factors in Endothelial Cells. Adv Healthc Mater 2015; 4:2056-2063. [PMID: 26270789 DOI: 10.1002/adhm.201500338] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/25/2015] [Indexed: 01/08/2023]
Abstract
Angiogenesis, the formation of blood vessels from pre-existing ones, is of vital importance during the early stages of bone healing. Extracellular stiffness plays an important role in regulating endothelial cell behavior and angiogenesis, but how this mechanical cue affects proliferation kinetics, gene regulation, and the expression of proteins implicated in angiogenesis and bone regeneration remains unclear. Using collagen-coated polyacrylamide (PAAm) hydrogels, human umbilical vein endothelial cells (HUVECs) are exposed to an environment that mimics the elastic properties of collagenous bone, and cellular proliferation and gene and protein expressions are assessed. The proliferation and gene expression of HUVECs are not differentially affected by culture on 3 or 30 kPa PAAm hydrogels, henceforth referred to as low and high stiffness gels, respectively. Although the proliferation and gene transcript levels remain unchanged, significant differences are found in the expressions of functional proteins and growth factors implicated both in the angiogenic and osteogenic processes. The down-regulation of the vascular endothelial growth factor receptor-2 protein with concomitant over-expression of caveolin-1, wingless-type 2, bone morphogenic protein 2, and basic fibroblast growth factor on the high stiffness PAAm hydrogel suggests that rigidity has a pro-angiogenic effect with inherent benefits for bone regeneration.
Collapse
Affiliation(s)
- Lívia Santos
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| | - Gregor Fuhrmann
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| | - Maya Juenet
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| | - Nadav Amdursky
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| | - Christine-Maria Horejs
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| | - Paola Campagnolo
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| | - Molly M. Stevens
- Department of Materials; Imperial College London; London SW7 2AZ UK
- Department of Bioengineering; Imperial College London; London SW7 2AZ UK
- Institute of Biomedical Engineering; Imperial College London; London SW7 2AZ UK
| |
Collapse
|
490
|
Caporizzo MA, Roco CM, Ferrer MCC, Grady ME, Parrish E, Eckmann DM, Composto RJ. Strain-rate Dependence of Elastic Modulus Reveals Silver Nanoparticle Induced Cytotoxicity. Nanobiomedicine (Rij) 2015; 2. [PMID: 26834855 PMCID: PMC4732735 DOI: 10.5772/61328] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Force-displacement measurements are taken at different rates with an atomic force microscope to assess the correlation between cell health and cell viscoelasticity in THP-1 cells that have been treated with a novel drug carrier. A variable indentation-rate viscoelastic analysis, VIVA, is employed to identify the relaxation time of the cells that are known to exhibit a frequency dependent stiffness. The VIVA agrees with a fluorescent viability assay. This indicates that dextran-lysozyme drug carriers are biocompatible and deliver concentrated toxic material (rhodamine or silver nanoparticles) to the cytoplasm of THP-1 cells. By modelling the frequency dependence of the elastic modulus, the VIVA provides three metrics of cytoplasmic viscoelasticity: a low frequency modulus, a high frequency modulus and viscosity. The signature of cytotoxicity by rhodamine or silver exposure is a frequency independent twofold increase in the elastic modulus and cytoplasmic viscosity, while the cytoskeletal relaxation time remains unchanged. This is consistent with the known toxic mechanism of silver nanoparticles, where metabolic stress causes an increase in the rigidity of the cytoplasm. A variable indentation-rate viscoelastic analysis is presented as a straightforward method to promote the self-consistent comparison between cells. This is paramount to the development of early diagnosis and treatment of disease.
Collapse
Affiliation(s)
| | - Charles M Roco
- Department of Materials Science Engineering, University of Pennsylvania, Pennsylvania, USA
| | - Maria Carme Coll Ferrer
- Department of Materials Science Engineering, University of Pennsylvania, Pennsylvania, USA; Department of Anesthesiology and Critical Care, University of Pennsylvania, Pennsylvania, USA
| | - Martha E Grady
- Department of Materials Science Engineering, University of Pennsylvania, Pennsylvania, USA; Department of Anesthesiology and Critical Care, University of Pennsylvania, Pennsylvania, USA
| | - Emmabeth Parrish
- Department of Materials Science Engineering, University of Pennsylvania, Pennsylvania, USA
| | - David M Eckmann
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Pennsylvania, USA
| | - Russell John Composto
- Department of Materials Science Engineering, University of Pennsylvania, Pennsylvania, USA
| |
Collapse
|
491
|
Regulation of tissue ingrowth into proteolytically degradable hydrogels. Acta Biomater 2015; 24:44-52. [PMID: 26079677 DOI: 10.1016/j.actbio.2015.06.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/23/2015] [Accepted: 06/08/2015] [Indexed: 12/28/2022]
Abstract
Regulation of the rate of cell ingrowth into and within a matrix is desirable for efficient tissue regeneration. Polyethylene glycol hydrogels crosslinked with matrix metalloproteinase (MMP) susceptible peptide sequences permit cell-controlled invasion. In this study, hydrogels of the same stiffness polymerised using different ratios of a readily degradable MMP peptide sequence (PAN-MMP) and a MMP peptide with a limited degradation capacity (MMP-9) were assessed both in vitro and in vivo for cellular invasion. The degree of invasion into the various hydrogels was found to be tightly linked to the relative proportion of each peptide both in vitro and in vivo. Furthermore a good correlation between in vitro and in vivo ingrowth was observed. These findings demonstrate a highly tunable model for regulating cellular invasion which is readily translatable to in vivo models. This finding may allow for further optimisation of aspects of regenerative scaffolds such as tissue invasion, growth factor release and cellular encapsulation. STATEMENT OF SIGNIFICANCE Degradable hydrogels are used in a wide range of tissue regeneration approaches. A particularly advantageous variant of these hydrogels is where due to peptide based crosslinking of the polymeric hydrogels, cell invasion rate is dependent on cellular enzymatic activity. This present study demonstrates a further refinement whereby both cellular and tissue invasion rates are finely regulated through the polymerisation of a hydrogel with varying combinations of enzymatically degradable peptides. Importantly this allows for invasion rates to be controlled without altering the biomechanical properties of the hydrogel such as stiffness. The latter can further influence cellular behaviour thus potentially interfering with the desired outcome.
Collapse
|
492
|
Lacraz G, Rouleau AJ, Couture V, Söllrald T, Drouin G, Veillette N, Grandbois M, Grenier G. Increased Stiffness in Aged Skeletal Muscle Impairs Muscle Progenitor Cell Proliferative Activity. PLoS One 2015; 10:e0136217. [PMID: 26295702 PMCID: PMC4546553 DOI: 10.1371/journal.pone.0136217] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/31/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Skeletal muscle aging is associated with a decreased regenerative potential due to the loss of function of endogenous stem cells or myogenic progenitor cells (MPCs). Aged skeletal muscle is characterized by the deposition of extracellular matrix (ECM), which in turn influences the biomechanical properties of myofibers by increasing their stiffness. Since the stiffness of the MPC microenvironment directly impacts MPC function, we hypothesized that the increase in muscle stiffness that occurs with aging impairs the behavior of MPCs, ultimately leading to a decrease in regenerative potential. RESULTS We showed that freshly isolated individual myofibers from aged mouse muscles contain fewer MPCs overall than myofibers from adult muscles, with fewer quiescent MPCs and more proliferative and differentiating MPCs. We observed alterations in cultured MPC behavior in aged animals, where the proliferation and differentiation of MPCs were lower and higher, respectively. These alterations were not linked to the intrinsic properties of aged myofibers, as shown by the similar values for the cumulative population-doubling values and fusion indexes. However, atomic force microscopy (AFM) indentation experiments revealed a nearly 4-fold increase in the stiffness of the MPC microenvironment. We further showed that the increase in stiffness is associated with alterations to muscle ECM, including the accumulation of collagen, which was correlated with higher hydroxyproline and advanced glycation end-product content. Lastly, we recapitulated the impaired MPC behavior observed in aging using a hydrogel substrate that mimics the stiffness of myofibers. CONCLUSIONS These findings provide novel evidence that the low regenerative potential of aged skeletal muscle is independent of intrinsic MPC properties but is related to the increase in the stiffness of the MPC microenvironment.
Collapse
Affiliation(s)
- Grégory Lacraz
- Centre Hospitalier de l’Université de Sherbrooke Research Center (CRCHUS), Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - André-Jean Rouleau
- Centre Hospitalier de l’Université de Sherbrooke Research Center (CRCHUS), Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Vanessa Couture
- Centre Hospitalier de l’Université de Sherbrooke Research Center (CRCHUS), Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Thomas Söllrald
- Department of Electrical and Computer Engineering, Faculty of Engineering, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Geneviève Drouin
- Centre Hospitalier de l’Université de Sherbrooke Research Center (CRCHUS), Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Noémie Veillette
- Centre Hospitalier de l’Université de Sherbrooke Research Center (CRCHUS), Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Michel Grandbois
- Centre Hospitalier de l’Université de Sherbrooke Research Center (CRCHUS), Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Department of Pharmacology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Guillaume Grenier
- Centre Hospitalier de l’Université de Sherbrooke Research Center (CRCHUS), Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Department of Orthopedic Surgery, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- * E-mail:
| |
Collapse
|
493
|
Hwang JH, Byun MR, Kim AR, Kim KM, Cho HJ, Lee YH, Kim J, Jeong MG, Hwang ES, Hong JH. Extracellular Matrix Stiffness Regulates Osteogenic Differentiation through MAPK Activation. PLoS One 2015; 10:e0135519. [PMID: 26262877 PMCID: PMC4532446 DOI: 10.1371/journal.pone.0135519] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 07/22/2015] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stem cell (MSC) differentiation is regulated by the extracellular matrix (ECM) through activation of intracellular signaling mediators. The stiffness of the ECM was shown to be an important regulatory factor for MSC differentiation, and transcriptional coactivator with PDZ-binding motif (TAZ) was identified as an effector protein for MSC differentiation. However, the detailed underlying mechanism regarding the role of ECM stiffness and TAZ in MSC differentiation is not yet fully understood. In this report, we showed that ECM stiffness regulates MSC fate through ERK or JNK activation. Specifically, a stiff hydrogel matrix stimulates osteogenic differentiation concomitant with increased nuclear localization of TAZ, but inhibits adipogenic differentiation. ERK and JNK activity was significantly increased in cells cultured on a stiff hydrogel. TAZ activation was induced by ERK or JNK activation on a stiff hydrogel because exposure to an ERK or JNK inhibitor significantly decreased the nuclear localization of TAZ, indicating that ECM stiffness-induced ERK or JNK activation is important for TAZ-driven osteogenic differentiation. Taken together, these results suggest that ECM stiffness regulates MSC differentiation through ERK or JNK activation.
Collapse
Affiliation(s)
- Jun-Ha Hwang
- Department of Life Sciences, School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Mi Ran Byun
- Department of Life Sciences, School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - A. Rum Kim
- Department of Life Sciences, School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Kyung Min Kim
- Department of Life Sciences, School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Hang Jun Cho
- Department of Life Sciences, School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Yo Han Lee
- Department of Life Sciences, School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Juwon Kim
- Department of Life Sciences, School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Mi Gyeong Jeong
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| | - Eun Sook Hwang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
- * E-mail: (J-H Hong); (ESH)
| | - Jeong-Ho Hong
- Department of Life Sciences, School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
- * E-mail: (J-H Hong); (ESH)
| |
Collapse
|
494
|
Thomas DG, Yenepalli A, Denais CM, Rape A, Beach JR, Wang YL, Schiemann WP, Baskaran H, Lammerding J, Egelhoff TT. Non-muscle myosin IIB is critical for nuclear translocation during 3D invasion. J Cell Biol 2015; 210:583-94. [PMID: 26261182 PMCID: PMC4539979 DOI: 10.1083/jcb.201502039] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/01/2015] [Indexed: 01/22/2023] Open
Abstract
Non-muscle myosin II (NMII) is reported to play multiple roles during cell migration and invasion. However, the exact biophysical roles of different NMII isoforms during these processes remain poorly understood. We analyzed the contributions of NMIIA and NMIIB in three-dimensional (3D) migration and in generating the forces required for efficient invasion by mammary gland carcinoma cells. Using traction force microscopy and microfluidic invasion devices, we demonstrated that NMIIA is critical for generating force during active protrusion, and NMIIB plays a major role in applying force on the nucleus to facilitate nuclear translocation through tight spaces. We further demonstrate that the nuclear membrane protein nesprin-2 is a possible linker coupling NMIIB-based force generation to nuclear translocation. Together, these data reveal a central biophysical role for NMIIB in nuclear translocation during 3D invasive migration, a result with relevance not only to cancer metastasis but for 3D migration in other settings such as embryonic cell migration and wound healing.
Collapse
Affiliation(s)
- Dustin G Thomas
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 441195 Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195
| | - Aishwarya Yenepalli
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 441195
| | - Celine Marie Denais
- Department of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853
| | - Andrew Rape
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720
| | - Jordan R Beach
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Yu-Li Wang
- Department of Biomedical Engineering, Carnegie Melon University, Pittsburgh, PA 15219
| | - William P Schiemann
- General Medical Sciences-Oncology, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Harihara Baskaran
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106
| | - Jan Lammerding
- Department of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853
| | - Thomas T Egelhoff
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 441195 Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195
| |
Collapse
|
495
|
RalB regulates contractility-driven cancer dissemination upon TGFβ stimulation via the RhoGEF GEF-H1. Sci Rep 2015; 5:11759. [PMID: 26152517 PMCID: PMC4495419 DOI: 10.1038/srep11759] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/03/2015] [Indexed: 01/18/2023] Open
Abstract
RalA and RalB proteins are key mediators of oncogenic Ras signaling in human oncogenesis. Herein we investigated the mechanistic contribution of Ral proteins to invasion of lung cancer A549 cells after induction of epithelial-mesenchymal transition (EMT) with TGFβ. We show that TGFβ-induced EMT promotes dissemination of A549 cells in a 2/3D assay, independently of proteolysis, by activating the Rho/ROCK pathway which generates actomyosin-dependent contractility forces that actively remodel the extracellular matrix, as assessed by Traction Force microscopy. RalB, but not RalA, is required for matrix deformation and cell dissemination acting via the RhoGEF GEF-H1, which associates with the Exocyst complex, a major Ral effector. Indeed, uncoupling of the Exocyst subunit Sec5 from GEF-H1 impairs RhoA activation, generation of traction forces and cell dissemination. These results provide a novel molecular mechanism underlying the control of cell invasion by RalB via a cross-talk with the Rho pathway.
Collapse
|
496
|
Adaptive rheology and ordering of cell cytoskeleton govern matrix rigidity sensing. Nat Commun 2015; 6:7525. [PMID: 26109233 PMCID: PMC4599139 DOI: 10.1038/ncomms8525] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 05/16/2015] [Indexed: 12/23/2022] Open
Abstract
Matrix rigidity sensing regulates a large variety of cellular processes and has important implications for tissue development and disease. However, how cells probe matrix rigidity, and hence respond to it, remains unclear. Here, we show that rigidity sensing and adaptation emerge naturally from actin cytoskeleton remodeling. Our in vitro experiments and theoretical modeling demonstrate a bi-phasic rheology of the actin cytoskeleton, which transitions from fluid on soft substrates to solid on stiffer ones. Furthermore, we find that increasing substrate stiffness correlates with the emergence of an orientational order in actin stress fibers, which exhibit an isotropic to nematic transition that we characterize quantitatively in the framework of active matter theory. These findings imply mechanisms mediated by a large-scale reinforcement of actin structures under stress, which could be the mechanical drivers of substrate stiffness dependent cell shape changes and cell polarity.
Collapse
|
497
|
Ali MY, Anand SV, Tangella K, Ramkumar D, Saif TA. Isolation of Primary Human Colon Tumor Cells from Surgical Tissues and Culturing Them Directly on Soft Elastic Substrates for Traction Cytometry. J Vis Exp 2015:e52532. [PMID: 26065530 DOI: 10.3791/52532] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Cancer cells respond to matrix mechanical stiffness in a complex manner using a coordinated, hierarchical mechano-chemical system composed of adhesion receptors and associated signal transduction membrane proteins, the cytoskeletal architecture, and molecular motors. Mechanosensitivity of different cancer cells in vitro are investigated primarily with immortalized cell lines or murine derived primary cells, not with primary human cancer cells. Hence, little is known about the mechanosensitivity of primary human colon cancer cells in vitro. Here, an optimized protocol is developed that describes the isolation of primary human colon cells from healthy and cancerous surgical human tissue samples. Isolated colon cells are then successfully cultured on soft (2 kPa stiffness) and stiff (10 kPa stiffness) polyacrylamide hydrogels and rigid polystyrene (~3.6 GPa stiffness) substrates functionalized by an extracellular matrix (fibronectin in this case). Fluorescent microbeads are embedded in soft gels near the cell culture surface, and traction assay is performed to assess cellular contractile stresses using free open access software. In addition, immunofluorescence microscopy on different stiffness substrates provides useful information about primary cell morphology, cytoskeleton organization and vinculin containing focal adhesions as a function of substrate rigidity.
Collapse
Affiliation(s)
- M Yakut Ali
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign
| | - Sandeep V Anand
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign
| | - Krishnarao Tangella
- College of Medicine, University of Illinois at Urbana-Champaign; Provena Covenant Medical Centre
| | - Davendra Ramkumar
- College of Medicine, University of Illinois at Urbana-Champaign; Provena Covenant Medical Centre
| | - Taher A Saif
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign; Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign;
| |
Collapse
|
498
|
Kee MF, Myers DR, Sakurai Y, Lam WA, Qiu Y. Platelet mechanosensing of collagen matrices. PLoS One 2015; 10:e0126624. [PMID: 25915413 PMCID: PMC4411076 DOI: 10.1371/journal.pone.0126624] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 04/03/2015] [Indexed: 11/18/2022] Open
Abstract
During vascular injury, platelets adhere to exposed subendothelial proteins, such as collagen, on the blood vessel walls to trigger clot formation. Although the biochemical signalings of platelet-collagen interactions have been well characterized, little is known about the role microenvironmental biomechanical properties, such as vascular wall stiffness, may have on clot formation. To that end, we investigated how substrates of varying stiffness conjugated with the same concentration of Type I collagen affect platelet adhesion, spreading, and activation. Using collagen-conjugated polyacrylamide (PA) gels of different stiffnesses, we observed that platelets do in fact mechanotransduce the stiffness cues of collagen substrates, manifesting in increased platelet spreading on stiffer substrates. In addition, increasing substrate stiffness also increases phosphatidylserine exposure, a key aspect of platelet activation that initiates coagulation on the platelet surface. Mechanistically, these collagen substrate stiffness effects are mediated by extracellular calcium levels and actomyosin pathways driven by myosin light chain kinase but not Rho-associated protein kinase. Overall, our results improve our understanding of how the mechanics of different tissues and stroma affect clot formation, what role the increased vessel wall stiffness in atherosclerosis may directly have on thrombosis leading to heart attacks and strokes, and how age-related increased vessel wall stiffness affects hemostasis and thrombosis.
Collapse
Affiliation(s)
- Matthew F. Kee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States of America
| | - David R. Myers
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States of America
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Winship Cancer Institute of Emory University, Atlanta, Georgia, United States of America
| | - Yumiko Sakurai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States of America
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Winship Cancer Institute of Emory University, Atlanta, Georgia, United States of America
| | - Wilbur A. Lam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States of America
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Winship Cancer Institute of Emory University, Atlanta, Georgia, United States of America
- Parker H. Petit Institute of Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Yongzhi Qiu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States of America
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Winship Cancer Institute of Emory University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
499
|
Fitzgerald MM, Bootsma K, Berberich JA, Sparks JL. Tunable stress relaxation behavior of an alginate-polyacrylamide hydrogel: comparison with muscle tissue. Biomacromolecules 2015; 16:1497-505. [PMID: 25812913 DOI: 10.1021/bm501845j] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Factors controlling the time-dependent mechanical properties of interpenetrating network (IPN) hydrogel materials are not well understood. In this study, alginate-polyacrylamide IPN were synthesized to mimic the stress relaxation behavior and elastic modulus of porcine muscle tissue. Hydrogel samples were created with single-parameter chemical concentration variations from a baseline formula to establish trends. The concentration of total monomer material had the largest effect on the elastic modulus, while concentration of the acrylamide cross-linker, N,N-methylenebis(acrylamide) (MBAA), changed the stress relaxation behavior most effectively. The IPN material was then tuned to mimic the mechanical response of muscle tissue using these trends. Swelling the hydrogel samples to equilibrium resulted in a dramatic decrease in both elastic modulus and stress relaxation behavior. Collectively, the results demonstrate that alginate-polyacrylamide IPN hydrogels can be tuned to closely mimic both the elastic and the viscoelastic behaviors of muscle tissue, although swelling detrimentally affects these desired properties.
Collapse
Affiliation(s)
- Martha M Fitzgerald
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, Ohio 45056, United States
| | - Katherine Bootsma
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, Ohio 45056, United States
| | - Jason A Berberich
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, Ohio 45056, United States
| | - Jessica L Sparks
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, Ohio 45056, United States
| |
Collapse
|
500
|
Sun J, Xiao Y, Wang S, Slepian MJ, Wong PK. Advances in Techniques for Probing Mechanoregulation of Tissue Morphogenesis. ACTA ACUST UNITED AC 2015; 20:127-37. [DOI: 10.1177/2211068214554802] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|