251
|
Li H, Duann P, Lin PH, Zhao L, Fan Z, Tan T, Zhou X, Sun M, Fu M, Orange M, Sermersheim M, Ma H, He D, Steinberg SM, Higgins R, Zhu H, John E, Zeng C, Guan J, Ma J. Modulation of wound healing and scar formation by MG53 protein-mediated cell membrane repair. J Biol Chem 2015; 290:24592-603. [PMID: 26306047 DOI: 10.1074/jbc.m115.680074] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Indexed: 01/08/2023] Open
Abstract
Cell membrane repair is an important aspect of physiology, and disruption of this process can result in pathophysiology in a number of different tissues, including wound healing, chronic ulcer and scarring. We have previously identified a novel tripartite motif family protein, MG53, as an essential component of the cell membrane repair machinery. Here we report the functional role of MG53 in the modulation of wound healing and scarring. Although MG53 is absent from keratinocytes and fibroblasts, remarkable defects in skin architecture and collagen overproduction are observed in mg53(-/-) mice, and these animals display delayed wound healing and abnormal scarring. Recombinant human MG53 (rhMG53) protein, encapsulated in a hydrogel formulation, facilitates wound healing and prevents scarring in rodent models of dermal injuries. An in vitro study shows that rhMG53 protects against acute injury to keratinocytes and facilitates the migration of fibroblasts in response to scratch wounding. During fibrotic remodeling, rhMG53 interferes with TGF-β-dependent activation of myofibroblast differentiation. The resulting down-regulation of α smooth muscle actin and extracellular matrix proteins contributes to reduced scarring. Overall, these studies establish a trifunctional role for MG53 as a facilitator of rapid injury repair, a mediator of cell migration, and a modulator of myofibroblast differentiation during wound healing. Targeting the functional interaction between MG53 and TGF-β signaling may present a potentially effective means for promoting scarless wound healing.
Collapse
Affiliation(s)
- Haichang Li
- From the Department of Surgery, Davis Heart and Lung Research Institute,
| | - Pu Duann
- From the Department of Surgery, Davis Heart and Lung Research Institute
| | - Pei-Hui Lin
- From the Department of Surgery, Davis Heart and Lung Research Institute
| | - Li Zhao
- From the Department of Surgery, Davis Heart and Lung Research Institute, the Division of Protein Therapeutics, TRIM-edicine Inc., Columbus, Ohio 43212
| | - Zhaobo Fan
- Department of Materials Science and Engineering, Ohio State University, Columbus, Ohio 43210
| | - Tao Tan
- From the Department of Surgery, Davis Heart and Lung Research Institute, the Division of Protein Therapeutics, TRIM-edicine Inc., Columbus, Ohio 43212
| | - Xinyu Zhou
- From the Department of Surgery, Davis Heart and Lung Research Institute
| | - Mingzhai Sun
- From the Department of Surgery, Davis Heart and Lung Research Institute
| | - Minghuan Fu
- Department of Materials Science and Engineering, Ohio State University, Columbus, Ohio 43210
| | - Matthew Orange
- the Department of Physical Education and Human Performance, Central Connecticut State University, New Britain, Connecticut 06050
| | | | - Hanley Ma
- From the Department of Surgery, Davis Heart and Lung Research Institute, Olentangy Liberty High School, Powell, Ohio 43065, and
| | - Duofen He
- the Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, China
| | | | - Robert Higgins
- From the Department of Surgery, Davis Heart and Lung Research Institute
| | - Hua Zhu
- From the Department of Surgery, Davis Heart and Lung Research Institute
| | - Elizabeth John
- the Division of Protein Therapeutics, TRIM-edicine Inc., Columbus, Ohio 43212
| | - Chunyu Zeng
- the Department of Cardiology, Chongqing Institute of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, China
| | - Jianjun Guan
- Department of Materials Science and Engineering, Ohio State University, Columbus, Ohio 43210
| | - Jianjie Ma
- From the Department of Surgery, Davis Heart and Lung Research Institute, the Division of Protein Therapeutics, TRIM-edicine Inc., Columbus, Ohio 43212,
| |
Collapse
|
252
|
Marchesano V, Gennari O, Mecozzi L, Grilli S, Ferraro P. Effects of Lithium Niobate Polarization on Cell Adhesion and Morphology. ACS APPLIED MATERIALS & INTERFACES 2015. [PMID: 26222955 DOI: 10.1021/acsami.5b05340] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Understanding how the interfacial effects influence cell adhesion and morphology is of fundamental interest for controlling function, growth, and movement of cells in vitro and in vivo. In particular, the influence of surface charges is well-known but still controversial, especially when new functional materials and methods are introduced. Here, the influence of the spontaneous polarization of ferroelectric lithium niobate (LN) on the adhesion properties of fibroblast cells is investigated. The spontaneous polarization of LN has one of the largest known magnitudes at room temperature (∼78 μC/cm(2)), and its orientation can be patterned easily by an external voltage, this motivating highly the investigation of its interaction with cells. Immunofluorescence and migration assays show strong evidence that the surface polarity regulates the adhesion functions, with enhanced spreading of the cytoskeleton on the negative face. The results suggest the potential of LN as a platform for investigating the role of charges on cellular processes, thus favoring new strategies in fabricating those biocompatible constructs used for tissue engineering. In fact, the orientation of the high-magnitude polarization can be patterned easily and, in combination with piezoelectric, pyroelectric, and photorefractive properties, may open the route to more sophisticated charge templates for modulating the cell response.
Collapse
Affiliation(s)
| | | | - Laura Mecozzi
- ‡University "Federico II", P. le Tecchio 80, 80125 Napoli, Italy
| | | | | |
Collapse
|
253
|
Paluch EK, Nelson CM, Biais N, Fabry B, Moeller J, Pruitt BL, Wollnik C, Kudryasheva G, Rehfeldt F, Federle W. Mechanotransduction: use the force(s). BMC Biol 2015; 13:47. [PMID: 26141078 PMCID: PMC4491211 DOI: 10.1186/s12915-015-0150-4] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mechanotransduction - how cells sense physical forces and translate them into biochemical and biological responses - is a vibrant and rapidly-progressing field, and is important for a broad range of biological phenomena. This forum explores the role of mechanotransduction in a variety of cellular activities and highlights intriguing questions that deserve further attention.
Collapse
Affiliation(s)
- Ewa K Paluch
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| | - Celeste M Nelson
- Chemical & Biological Engineering, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ, 08544, USA
| | - Nicolas Biais
- Biology Department, Brooklyn College and the Graduate Center of the City University of New York, 2900 Bedford avenue, Brooklyn, NY, 11210, USA
| | - Ben Fabry
- Department of Physics, University of Erlangen-Nuremberg, Henkestrasse 91, 91052, Erlangen, Germany
| | - Jens Moeller
- Department of Mechanical Engineering, Microsystems Laboratory, Stanford University, 496 Lomita Mall, Durand Building Rm 102, Stanford, CA, 94305, USA
| | - Beth L Pruitt
- Department of Mechanical Engineering and Molecular and Cellular Physiology, Microsystems Laboratory, Stanford University, by courtesy, 496 Lomita Mall, Durand Building Rm 213, Stanford, CA, 94305, USA
| | - Carina Wollnik
- Georg-August-University, 3rd Institute of Physics - Biophysics, Friedrich-Hund-Platz 1, 37077, Göttingen, Germany
| | - Galina Kudryasheva
- Georg-August-University, 3rd Institute of Physics - Biophysics, Friedrich-Hund-Platz 1, 37077, Göttingen, Germany
| | - Florian Rehfeldt
- Georg-August-University, 3rd Institute of Physics - Biophysics, Friedrich-Hund-Platz 1, 37077, Göttingen, Germany
| | - Walter Federle
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK
| |
Collapse
|
254
|
Ghosh S, Kollar B, Nahar T, Suresh Babu S, Wojtowicz A, Sticht C, Gretz N, Wagner AH, Korff T, Hecker M. Loss of the mechanotransducer zyxin promotes a synthetic phenotype of vascular smooth muscle cells. J Am Heart Assoc 2015; 4:e001712. [PMID: 26071033 PMCID: PMC4599528 DOI: 10.1161/jaha.114.001712] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Background Exposure of vascular smooth muscle cells (VSMCs) to excessive cyclic stretch such as in hypertension causes a shift in their phenotype. The focal adhesion protein zyxin can transduce such biomechanical stimuli to the nucleus of both endothelial cells and VSMCs, albeit with different thresholds and kinetics. However, there is no distinct vascular phenotype in young zyxin-deficient mice, possibly due to functional redundancy among other gene products belonging to the zyxin family. Analyzing zyxin function in VSMCs at the cellular level might thus offer a better mechanistic insight. We aimed to characterize zyxin-dependent changes in gene expression in VSMCs exposed to biomechanical stretch and define the functional role of zyxin in controlling the resultant VSMC phenotype. Methods and Results DNA microarray analysis was used to identify genes and pathways that were zyxin regulated in static and stretched human umbilical artery–derived and mouse aortic VSMCs. Zyxin-null VSMCs showed a remarkable shift to a growth-promoting, less apoptotic, promigratory and poorly contractile phenotype with ≈90% of the stretch-responsive genes being zyxin dependent. Interestingly, zyxin-null cells already seemed primed for such a synthetic phenotype, with mechanical stretch further accentuating it. This could be accounted for by higher RhoA activity and myocardin-related transcription factor-A mainly localized to the nucleus of zyxin-null VSMCs, and a condensed and localized accumulation of F-actin upon stretch. Conclusions At the cellular level, zyxin is a key regulator of stretch-induced gene expression. Loss of zyxin drives VSMCs toward a synthetic phenotype, a process further consolidated by exaggerated stretch.
Collapse
Affiliation(s)
- Subhajit Ghosh
- Institute of Physiology and Pathophysiology, University of Heidelberg, Germany (S.G., B.K., T.N., A.H.W., T.K.)
| | - Branislav Kollar
- Institute of Physiology and Pathophysiology, University of Heidelberg, Germany (S.G., B.K., T.N., A.H.W., T.K.)
| | - Taslima Nahar
- Institute of Physiology and Pathophysiology, University of Heidelberg, Germany (S.G., B.K., T.N., A.H.W., T.K.)
| | - Sahana Suresh Babu
- Department of Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX (S.S.B.)
| | - Agnieszka Wojtowicz
- Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland (A.W.)
| | - Carsten Sticht
- ZMF, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.S., N.G.)
| | - Norbert Gretz
- ZMF, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany (C.S., N.G.)
| | - Andreas H Wagner
- Institute of Physiology and Pathophysiology, University of Heidelberg, Germany (S.G., B.K., T.N., A.H.W., T.K.)
| | - Thomas Korff
- Institute of Physiology and Pathophysiology, University of Heidelberg, Germany (S.G., B.K., T.N., A.H.W., T.K.)
| | - Markus Hecker
- Institute of Physiology and Pathophysiology, University of Heidelberg and Deutsches Zentrum Für Herz-Kreislauf-Forschung E.V. (DZHK), Partner site Heidelberg/Mannheim, Germany (M.H.)
| |
Collapse
|
255
|
Eltzner B, Wollnik C, Gottschlich C, Huckemann S, Rehfeldt F. The filament sensor for near real-time detection of cytoskeletal fiber structures. PLoS One 2015; 10:e0126346. [PMID: 25996921 PMCID: PMC4440737 DOI: 10.1371/journal.pone.0126346] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 04/01/2015] [Indexed: 12/18/2022] Open
Abstract
A reliable extraction of filament data from microscopic images is of high interest in the analysis of acto-myosin structures as early morphological markers in mechanically guided differentiation of human mesenchymal stem cells and the understanding of the underlying fiber arrangement processes. In this paper, we propose the filament sensor (FS), a fast and robust processing sequence which detects and records location, orientation, length, and width for each single filament of an image, and thus allows for the above described analysis. The extraction of these features has previously not been possible with existing methods. We evaluate the performance of the proposed FS in terms of accuracy and speed in comparison to three existing methods with respect to their limited output. Further, we provide a benchmark dataset of real cell images along with filaments manually marked by a human expert as well as simulated benchmark images. The FS clearly outperforms existing methods in terms of computational runtime and filament extraction accuracy. The implementation of the FS and the benchmark database are available as open source.
Collapse
Affiliation(s)
- Benjamin Eltzner
- Institute for Mathematical Stochastics, Georg-August-University, 37077 Göttingen, Germany
| | - Carina Wollnik
- Third Institute of Physics-Biophysics, Georg-August-University, 37077 Göttingen, Germany
| | - Carsten Gottschlich
- Institute for Mathematical Stochastics, Georg-August-University, 37077 Göttingen, Germany
| | - Stephan Huckemann
- Institute for Mathematical Stochastics, Georg-August-University, 37077 Göttingen, Germany
| | - Florian Rehfeldt
- Third Institute of Physics-Biophysics, Georg-August-University, 37077 Göttingen, Germany
| |
Collapse
|
256
|
Adderley SP, Lawrence C, Madonia E, Olubadewo JO, Breslin JW. Histamine activates p38 MAP kinase and alters local lamellipodia dynamics, reducing endothelial barrier integrity and eliciting central movement of actin fibers. Am J Physiol Cell Physiol 2015; 309:C51-9. [PMID: 25948734 DOI: 10.1152/ajpcell.00096.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/03/2015] [Indexed: 11/22/2022]
Abstract
The role of the actin cytoskeleton in endothelial barrier function has been debated for nearly four decades. Our previous investigation revealed spontaneous local lamellipodia in confluent endothelial monolayers that appear to increase overlap at intercellular junctions. We tested the hypothesis that the barrier-disrupting agent histamine would reduce local lamellipodia protrusions and investigated the potential involvement of p38 mitogen-activated protein (MAP) kinase activation and actin stress fiber formation. Confluent monolayers of human umbilical vein endothelial cells (HUVEC) expressing green fluorescent protein-actin were studied using time-lapse fluorescence microscopy. The protrusion and withdrawal characteristics of local lamellipodia were assessed before and after addition of histamine. Changes in barrier function were determined using electrical cell-substrate impedance sensing. Histamine initially decreased barrier function, lamellipodia protrusion frequency, and lamellipodia protrusion distance. A longer time for lamellipodia withdrawal and reduced withdrawal distance and velocity accompanied barrier recovery. After barrier recovery, a significant number of cortical fibers migrated centrally, eventually resembling actin stress fibers. The p38 MAP kinase inhibitor SB203580 attenuated the histamine-induced decreases in barrier function and lamellipodia protrusion frequency. SB203580 also inhibited the histamine-induced decreases in withdrawal distance and velocity, and the subsequent actin fiber migration. These data suggest that histamine can reduce local lamellipodia protrusion activity through activation of p38 MAP kinase. The findings also suggest that local lamellipodia have a role in maintaining endothelial barrier integrity. Furthermore, we provide evidence that actin stress fiber formation may be a reaction to, rather than a cause of, reduced endothelial barrier integrity.
Collapse
Affiliation(s)
- Shaquria P Adderley
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida; and
| | - Curtis Lawrence
- Biology Unit, Department of Natural Sciences, Southern University at New Orleans, New Orleans, Louisiana
| | - Eyong Madonia
- Biology Unit, Department of Natural Sciences, Southern University at New Orleans, New Orleans, Louisiana
| | - Joseph O Olubadewo
- Biology Unit, Department of Natural Sciences, Southern University at New Orleans, New Orleans, Louisiana
| | - Jerome W Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida; and
| |
Collapse
|
257
|
Saito K, Shiino T, Kurihara H, Harita Y, Hattori S, Ohta Y. Afadin regulates RhoA/Rho-associated protein kinase signaling to control formation of actin stress fibers in kidney podocytes. Cytoskeleton (Hoboken) 2015; 72:146-56. [PMID: 25712270 DOI: 10.1002/cm.21211] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 01/19/2015] [Accepted: 02/20/2015] [Indexed: 11/05/2022]
Abstract
The function of kidney podocytes is closely associated with actin cytoskeleton. Rho family small GTPase RhoA promotes stress fiber assembly through Rho-associated protein kinase (ROCK)-dependent myosin II phosphorylation and plays an important role in maintenance of actin stress fibers of podocytes. However, little is known how stress fiber assembly is regulated in podocytes. Here, we found that afadin, an actin filament-binding protein, is required for RhoA/ROCK-dependent formation of actin stress fibers in rat podocyte C7 cells. We show that depletion of afadin in C7 cells induced loss of actin stress fibers. Conversely, forced expression of afadin increased the formation of actin stress fibers. Depletion of afadin inactivated RhoA and reduced the phosphorylation of myosin II. Moreover, the DIL domain of afadin appears to be responsible for actin stress fiber formation. Thus, afadin mediates RhoA/ROCK signaling and contributes to the formation of actin stress fibers in podocyte cells.
Collapse
Affiliation(s)
- Koji Saito
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, Kanagawa, Japan
| | | | | | | | | | | |
Collapse
|
258
|
Kassianidou E, Kumar S. A biomechanical perspective on stress fiber structure and function. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:3065-74. [PMID: 25896524 DOI: 10.1016/j.bbamcr.2015.04.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 04/05/2015] [Accepted: 04/08/2015] [Indexed: 01/11/2023]
Abstract
Stress fibers are actomyosin-based bundles whose structural and contractile properties underlie numerous cellular processes including adhesion, motility and mechanosensing. Recent advances in high-resolution live-cell imaging and single-cell force measurement have dramatically sharpened our understanding of the assembly, connectivity, and evolution of various specialized stress fiber subpopulations. This in turn has motivated interest in understanding how individual stress fibers generate tension and support cellular structure and force generation. In this review, we discuss approaches for measuring the mechanical properties of single stress fibers. We begin by discussing studies conducted in cell-free settings, including strategies based on isolation of intact stress fibers and reconstitution of stress fiber-like structures from purified components. We then discuss measurements obtained in living cells based both on inference of stress fiber properties from whole-cell mechanical measurements (e.g., atomic force microscopy) and on direct interrogation of single stress fibers (e.g., subcellular laser nanosurgery). We conclude by reviewing various mathematical models of stress fiber function that have been developed based on these experimental measurements. An important future challenge in this area will be the integration of these sophisticated biophysical measurements with the field's increasingly detailed molecular understanding of stress fiber assembly, dynamics, and signal transduction. This article is part of a Special Issue entitled: Mechanobiology.
Collapse
Affiliation(s)
- Elena Kassianidou
- Department of Bioengineering, University of California, Berkeley, United States
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, United States.
| |
Collapse
|
259
|
Abstract
The sense of touch informs us of the physical properties of our surroundings and is a critical aspect of communication. Before touches are perceived, mechanical signals are transmitted quickly and reliably from the skin's surface to mechano-electrical transduction channels embedded within specialized sensory neurons. We are just beginning to understand how soft tissues participate in force transmission and how they are deformed. Here, we review empirical and theoretical studies of single molecules and molecular ensembles thought to be involved in mechanotransmission and apply the concepts emerging from this work to the sense of touch. We focus on the nematode Caenorhabditis elegans as a well-studied model for touch sensation in which mechanics can be studied on the molecular, cellular, and systems level. Finally, we conclude that force transmission is an emergent property of macromolecular cellular structures that mutually stabilize one another.
Collapse
Affiliation(s)
- Michael Krieg
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alex Dunn
- Department of Chemical Engineering, Stanford University School of Engineering, Stanford, CA, USA
| | - Miriam B. Goodman
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
260
|
Undifferentiated bronchial fibroblasts derived from asthmatic patients display higher elastic modulus than their non-asthmatic counterparts. PLoS One 2015; 10:e0116840. [PMID: 25679502 PMCID: PMC4334506 DOI: 10.1371/journal.pone.0116840] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 12/15/2014] [Indexed: 12/31/2022] Open
Abstract
During asthma development, differentiation of epithelial cells and fibroblasts towards the contractile phenotype is associated with bronchial wall remodeling and airway constriction. Pathological fibroblast-to-myofibroblast transition (FMT) can be triggered by local inflammation of bronchial walls. Recently, we have demonstrated that human bronchial fibroblasts (HBFs) derived from asthmatic patients display some inherent features which facilitate their FMT in vitro. In spite of intensive research efforts, these properties remain unknown. Importantly, the role of undifferentiated HBFs in the asthmatic process was systematically omitted. Specifically, biomechanical properties of undifferentiated HBFs have not been considered in either FMT or airway remodeling in vivo. Here, we combine atomic force spectroscopy with fluorescence microscopy to compare mechanical properties and actin cytoskeleton architecture of HBFs derived from asthmatic patients and non-asthmatic donors. Our results demonstrate that asthmatic HBFs form thick and aligned ‘ventral’ stress fibers accompanied by enlarged focal adhesions. The differences in cytoskeleton architecture between asthmatic and non-asthmatic cells correlate with higher elastic modulus of asthmatic HBFs and their increased predilection to TGF-β-induced FMT. Due to the obvious links between cytoskeleton architecture and mechanical equilibrium, our observations indicate that HBFs derived from asthmatic bronchi can develop considerably higher static tension than non-asthmatic HBFs. This previously unexplored property of asthmatic HBFs may be potentially important for their myofibroblastic differentiation and bronchial wall remodeling during asthma development.
Collapse
|
261
|
Breslin JW, Zhang XE, Worthylake RA, Souza-Smith FM. Involvement of local lamellipodia in endothelial barrier function. PLoS One 2015; 10:e0117970. [PMID: 25658915 PMCID: PMC4320108 DOI: 10.1371/journal.pone.0117970] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 01/06/2015] [Indexed: 01/15/2023] Open
Abstract
Recently we observed that endothelial cells cultured in tightly confluent monolayers display frequent local lamellipodia, and that thrombin, an agent that increases endothelial permeability, reduces lamellipodia protrusions. This led us to test the hypothesis that local lamellipodia contribute to endothelial barrier function. Movements of subcellular structures containing GFP-actin or VE-cadherin-GFP expressed in endothelial cells were recorded using time-lapse microscopy. Transendothelial electrical resistance (TER) served as an index of endothelial barrier function. Changes in both lamellipodia dynamics and TER were assessed during baseline and after cells were treated with either the barrier-disrupting agent thrombin, or the barrier-stabilizing agent sphingosine-1-phosphate (S1P). The myosin II inhibitor blebbistatin was used to selectively block lamellipodia formation, and was used to test their role in the barrier function of endothelial cell monolayers and isolated, perfused rat mesenteric venules. Myosin light chain (MLC) phosphorylation was assessed by immunofluorescence microscopy. Rac1 and RhoA activation were evaluated using G-LISA assays. The role of Rac1 was tested with the specific inhibitor NSC23766 or by expressing wild-type or dominant negative GFP-Rac1. The results show that thrombin rapidly decreased both TER and the lamellipodia protrusion frequency. S1P rapidly increased TER in association with increased protrusion frequency. Blebbistatin nearly abolished local lamellipodia protrusions while cortical actin fibers and stress fibers remained intact. Blebbistatin also significantly decreased TER of cultured endothelial cells and increased permeability of isolated rat mesenteric venules. Both thrombin and S1P increased MLC phosphorylation and activation of RhoA. However, thrombin and S1P had differential impacts on Rac1, correlating with the changes in TER and lamellipodia protrusion frequency. Overexpression of Rac1 elevated, while NSC23766 and dominant negative Rac1 reduced barrier function and lamellipodia activity. Combined, these data suggest that local lamellipodia, driven by myosin II and Rac1, are important for dynamic changes in endothelial barrier integrity.
Collapse
Affiliation(s)
- Jerome W. Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
- * E-mail:
| | - Xun E. Zhang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Rebecca A. Worthylake
- Department of Pharmacology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Flavia M. Souza-Smith
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, United States of America
| |
Collapse
|
262
|
Wallert MA, Hammes D, Nguyen T, Kiefer L, Berthelsen N, Kern A, Anderson-Tiege K, Shabb JB, Muhonen WW, Grove BD, Provost JJ. RhoA Kinase (Rock) and p90 Ribosomal S6 Kinase (p90Rsk) phosphorylation of the sodium hydrogen exchanger (NHE1) is required for lysophosphatidic acid-induced transport, cytoskeletal organization and migration. Cell Signal 2015; 27:498-509. [PMID: 25578862 DOI: 10.1016/j.cellsig.2015.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 01/03/2015] [Indexed: 12/28/2022]
Abstract
The sodium hydrogen exchanger isoform one (NHE1) plays a critical role coordinating asymmetric events at the leading edge of migrating cells and is regulated by a number of phosphorylation events influencing both the ion transport and cytoskeletal anchoring required for directed migration. Lysophosphatidic acid (LPA) activation of RhoA kinase (Rock) and the Ras-ERK growth factor pathway induces cytoskeletal reorganization, activates NHE1 and induces an increase in cell motility. We report that both Rock I and II stoichiometrically phosphorylate NHE1 at threonine 653 in vitro using mass spectrometry and reconstituted kinase assays. In fibroblasts expressing NHE1 alanine mutants for either Rock (T653A) or ribosomal S6 kinase (Rsk; S703A) we show that each site is partially responsible for the LPA-induced increase in transport activity while NHE1 phosphorylation by either Rock or Rsk at their respective site is sufficient for LPA stimulated stress fiber formation and migration. Furthermore, mutation of either T653 or S703 leads to a higher basal pH level and a significantly higher proliferation rate. Our results identify the direct phosphorylation of NHE1 by Rock and suggest that both RhoA and Ras pathways mediate NHE1-dependent ion transport and migration in fibroblasts.
Collapse
Affiliation(s)
- Mark A Wallert
- Minnesota State University Moorhead, Department of Biosciences, Moorhead, MN, USA
| | - Daniel Hammes
- Minnesota State University Moorhead, Department of Biosciences, Moorhead, MN, USA
| | - Tony Nguyen
- Minnesota State University Moorhead, Department of Biosciences, Moorhead, MN, USA
| | - Lea Kiefer
- University of San Diego, Department of Chemistry and Biochemistry, San Diego, CA, USA
| | - Nick Berthelsen
- Minnesota State University Moorhead, Department of Biosciences, Moorhead, MN, USA
| | - Andrew Kern
- Minnesota State University Moorhead, Department of Biosciences, Moorhead, MN, USA
| | | | - John B Shabb
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, USA
| | - Wallace W Muhonen
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, USA
| | - Bryon D Grove
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, USA
| | - Joseph J Provost
- University of San Diego, Department of Chemistry and Biochemistry, San Diego, CA, USA.
| |
Collapse
|
263
|
Soon CF, Tee KS, Youseffi M, Denyer MCT. Tracking traction force changes of single cells on the liquid crystal surface. BIOSENSORS-BASEL 2015; 5:13-24. [PMID: 25808839 PMCID: PMC4384078 DOI: 10.3390/bios5010013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/02/2014] [Indexed: 11/16/2022]
Abstract
Cell migration is a key contributor to wound repair. This study presents findings indicating that the liquid crystal based cell traction force transducer (LCTFT) system can be used in conjunction with a bespoke cell traction force mapping (CTFM) software to monitor cell/surface traction forces from quiescent state in real time. In this study, time-lapse photo microscopy allowed cell induced deformations in liquid crystal coated substrates to be monitored and analyzed. The results indicated that the system could be used to monitor the generation of cell/surface forces in an initially quiescent cell, as it migrated over the culture substrate, via multiple points of contact between the cell and the surface. Future application of this system is the real-time assaying of the pharmacological effects of cytokines on the mechanics of cell migration.
Collapse
Affiliation(s)
- Chin Fhong Soon
- Biosensor and Bioengineering Laboratory, MiNT-SRC, Universiti Tun Hussein Onn Malaysia, 83000 Batu Pahat, Malaysia.
| | - Kian Sek Tee
- Biosensor and Bioengineering Laboratory, MiNT-SRC, Universiti Tun Hussein Onn Malaysia, 83000 Batu Pahat, Malaysia.
| | - Mansour Youseffi
- School of Engineering, Design and Technology-Medical Engineering, University of Bradford, BD7 1DP Bradford, UK.
| | - Morgan C T Denyer
- School of Medical Sciences, University of Bradford, BD7 1DP Bradford, UK.
| |
Collapse
|
264
|
Schenk M, Aykut B, Teske C, Giese NA, Weitz J, Welsch T. Salinomycin inhibits growth of pancreatic cancer and cancer cell migration by disruption of actin stress fiber integrity. Cancer Lett 2014; 358:161-169. [PMID: 25529011 DOI: 10.1016/j.canlet.2014.12.037] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 12/02/2014] [Accepted: 12/13/2014] [Indexed: 12/28/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by aggressive growth, early metastasis and high resistance to chemotherapy. Salinomycin is a promising compound eliminating cancer stem cells and retarding cancer cell migration. The present study investigated the effectiveness of salinomycin against PDAC in vivo and elucidated the mechanism of PDAC growth inhibition. Salinomycin treatment was well tolerated by the mice and significantly reduced tumor growth after 19 days compared to the control group (each n = 16). There was a trend that salinomycin also impeded metastatic spread to the liver and peritoneum. Whereas salinomycin moderately induced apoptosis and retarded proliferation at 5-10 µM, it strongly inhibited cancer cell migration that was accompanied by a marked loss of actin stress fibers after 6-9 h. Salinomycin silenced RhoA activity, and loss of stress fibers could be reversed by Rho activation. Moreover, salinomycin dislocated fascin from filopodia and stimulated Rac-associated circular dorsal ruffle formation. In conclusion, salinomycin is an effective and promising compound against PDAC. Besides its known stem cell-specific cytotoxic effects, salinomycin blocks cancer cell migration by disrupting stress fiber integrity and affecting the mutual Rho-GTPase balance.
Collapse
Affiliation(s)
- Miriam Schenk
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, 69120 Heidelberg, Germany
| | - Berk Aykut
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, 69120 Heidelberg, Germany
| | - Christian Teske
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Nathalia A Giese
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, 69120 Heidelberg, Germany
| | - Juergen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Germany
| | - Thilo Welsch
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, 69120 Heidelberg, Germany; Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Germany.
| |
Collapse
|
265
|
A computational study of stress fiber-focal adhesion dynamics governing cell contractility. Biophys J 2014; 106:1890-901. [PMID: 24806921 DOI: 10.1016/j.bpj.2014.03.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 03/02/2014] [Accepted: 03/06/2014] [Indexed: 01/12/2023] Open
Abstract
We apply a recently developed model of cytoskeletal force generation to study a cell's intrinsic contractility, as well as its response to external loading. The model is based on a nonequilibrium thermodynamic treatment of the mechanochemistry governing force in the stress fiber-focal adhesion system. Our computational study suggests that the mechanical coupling between the stress fibers and focal adhesions leads to a complex, dynamic, mechanochemical response. We collect the results in response maps whose regimes are distinguished by the initial geometry of the stress fiber-focal adhesion system, and by the external load on the cell. The results from our model connect qualitatively with recent studies on the force response of smooth muscle cells on arrays of polymeric microposts.
Collapse
|
266
|
Gill MB, Turner R, Stevenson PG, Way M. KSHV-TK is a tyrosine kinase that disrupts focal adhesions and induces Rho-mediated cell contraction. EMBO J 2014; 34:448-65. [PMID: 25471072 DOI: 10.15252/embj.201490358] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Paradoxically, the thymidine kinase (TK) encoded by Kaposi sarcoma-associated herpesvirus (KSHV) is an extremely inefficient nucleoside kinase, when compared to TKs from related herpesviruses. We now show that KSHV-TK, in contrast to HSV1-TK, associates with the actin cytoskeleton and induces extensive cell contraction followed by membrane blebbing. These dramatic changes in cell morphology depend on the auto-phosphorylation of tyrosines 65, 85 and 120 in the N-terminus of KSHV-TK. Phosphorylation of tyrosines 65/85 and 120 results in an interaction with Crk family proteins and the p85 regulatory subunit of PI3-Kinase, respectively. The interaction of Crk with KSHV-TK leads to tyrosine phoshorylation of this cellular adaptor. Auto-phosphorylation of KSHV-TK also induces a loss of FAK and paxillin from focal adhesions, resulting in activation of RhoA-ROCK signalling to myosin II and cell contraction. In the absence of FAK or paxillin, KSHV-TK has no effect on focal adhesion integrity or cell morphology. Our observations demonstrate that by acting as a tyrosine kinase, KSHV-TK modulates signalling and cell morphology.
Collapse
Affiliation(s)
- Michael B Gill
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Rachel Turner
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Philip G Stevenson
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Michael Way
- Cell Motility Laboratory, London Research Institute Cancer Research UK, London, UK
| |
Collapse
|
267
|
Xu X, Wang W, Kratz K, Fang L, Li Z, Kurtz A, Ma N, Lendlein A. Controlling major cellular processes of human mesenchymal stem cells using microwell structures. Adv Healthc Mater 2014; 3:1991-2003. [PMID: 25313500 DOI: 10.1002/adhm.201400415] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/02/2014] [Indexed: 01/17/2023]
Abstract
Directing stem cells towards a desired location and function by utilizing the structural cues of biomaterials is a promising approach for inducing effective tissue regeneration. Here, the cellular response of human adipose-derived mesenchymal stem cells (hADSCs) to structural signals from microstructured substrates comprising arrays of square-shaped or round-shaped microwells is explored as a transitional model between 2D and 3D systems. Microwells with a side length/diameter of 50 μm show advantages over 10 μm and 25 μm microwells for accommodating hADSCs within single microwells rather than in the inter-microwell area. The cell morphologies are three-dimensionally modulated by the microwell structure due to differences in focal adhesion and consequent alterations of the cytoskeleton. In contrast to the substrate with 50 μm round-shaped microwells, the substrate with 50 μm square-shaped microwells promotes the proliferation and osteogenic differentiation potential of hADSCs but reduces the cell migration velocity and distance. Such microwell shape-dependent modulatory effects are highly associated with Rho/ROCK signaling. Following ROCK inhibition, the differences in migration, proliferation, and osteogenesis between cells on different substrates are diminished. These results highlight the possibility to control stem cell functions through the use of structured microwells combined with the manipulation of Rho/ROCK signaling.
Collapse
Affiliation(s)
- Xun Xu
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Institute of Chemistry and Biochemistry; Freie Universität Berlin; Takustraße 3 14195 Berlin Germany
| | - Weiwei Wang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
| | - Karl Kratz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Helmholtz Virtual Institute −Multifunctional Materials in Medicine; Berlin and Teltow; Kantstraße 55 14513 Teltow Germany
| | - Liang Fang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
| | - Zhengdong Li
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Institute of Chemistry and Biochemistry; Freie Universität Berlin; Takustraße 3 14195 Berlin Germany
| | - Andreas Kurtz
- Berlin-Brandenburg Center for Regenerative Therapies; Charité - University Medicine Berlin; Augustenburger Platz 1 13353 Berlin Germany
- College of Veterinary Medicine and Research Institute for Veterinary Science; Seoul National University; Gwangk-ro 1 Gwanak-gu Seoul 151-747 Republic of Korea
| | - Nan Ma
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Institute of Chemistry and Biochemistry; Freie Universität Berlin; Takustraße 3 14195 Berlin Germany
- Helmholtz Virtual Institute −Multifunctional Materials in Medicine; Berlin and Teltow; Kantstraße 55 14513 Teltow Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Institute of Chemistry and Biochemistry; Freie Universität Berlin; Takustraße 3 14195 Berlin Germany
- Helmholtz Virtual Institute −Multifunctional Materials in Medicine; Berlin and Teltow; Kantstraße 55 14513 Teltow Germany
| |
Collapse
|
268
|
Kong X, Yan D, Sun J, Wu X, Mulder H, Hua X, Ma X. Glucagon-like peptide 1 stimulates insulin secretion via inhibiting RhoA/ROCK signaling and disassembling glucotoxicity-induced stress fibers. Endocrinology 2014; 155:4676-85. [PMID: 25243854 DOI: 10.1210/en.2014-1314] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Chronic hyperglycemia leads to pancreatic β-cell dysfunction characterized by diminished glucose-stimulated insulin secretion (GSIS), but the precise cellular processes involved are largely unknown. Here we show that pancreatic β-cells chronically exposed to a high glucose level displayed substantially increased amounts of stress fibers compared with β-cells cultured at a low glucose level. β-Cells at high glucose were refractory to glucose-induced actin cytoskeleton remodeling and insulin secretion. Importantly, F-actin depolymerization by either cytochalasin B or latrunculin B restored glucotoxicity-diminished GSIS. The effects of glucotoxicity on increasing stress fibers and reducing GSIS were reversed by Y-27632, a Rho-associated kinase (ROCK)-specific inhibitor, which caused actin depolymerization and enhanced GSIS. Notably, glucagon-like peptide-1-(7-36) amide (GLP-1), a peptide hormone that stimulates GSIS at both normal and hyperglycemic conditions, also reversed glucotoxicity-induced increase of stress fibers and reduction of GSIS. In addition, GLP-1 inhibited glucotoxicity-induced activation of RhoA/ROCK and thereby resulted in actin depolymerization and potentiation of GSIS. Furthermore, this effect of GLP-1 was mimicked by cAMP-increasing agents forskolin and 3-isobutyl-1-methylxanthine as well as the protein kinase A agonist 6-Bnz-cAMP-AM whereas it was abolished by the protein kinase A inhibitor Rp-Adenosine 3',5'-cyclic monophosphorothioate triethylammonium salt. To establish a clinical relevance of our findings, we examined the association of genetic variants of RhoA/ROCK with metabolic traits in homeostasis model assessment index of insulin resistance. Several single-nucleotide polymorphisms in and around RHOA were associated with elevated fasting insulin and homeostasis model assessment index of insulin resistance, suggesting a possible role in metabolic dysregulation. Collectively these findings unravel a novel mechanism whereby GLP-1 potentiates glucotoxicity-diminished GSIS by depolymerizing F-actin cytoskeleton via protein kinase A-mediated inhibition of the RhoA-ROCK signaling pathway.
Collapse
Affiliation(s)
- Xiangchen Kong
- Shenzhen University Diabetes Center (X.K., D.Y., X.W., X.H., X.M.) and Shenzhen University, Shenzhen 518060, People's Republic of China; Lund University Diabetes Centre (J.S., H.M.), Unit of Molecular Metabolism, SE-205 02 Malmö, Sweden; and University of Pennsylvania Perelman School of Medicine (X.H.), Philadelphia, Pennsylvania 19104
| | | | | | | | | | | | | |
Collapse
|
269
|
Hänisch J, Stradal TE, Rottner K. A novel contractility pathway operating in Salmonella invasion. Virulence 2014; 3:81-6. [DOI: 10.4161/viru.3.1.18454] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
270
|
Cheng CY, Mruk DD. Actin binding proteins and spermiogenesis: Some unexpected findings. SPERMATOGENESIS 2014; 1:99-104. [PMID: 22319657 DOI: 10.4161/spmg.1.2.16913] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 06/05/2011] [Accepted: 06/07/2011] [Indexed: 12/18/2022]
Abstract
Drebrin E, an actin-binding protein lacking intrinsic activity in the regulation of actin dynamics (e.g., polymerization, capping, nucleation, branching, cross-linking, bundling and severing), is known to recruit actin regulatory proteins to a specific cellular site. Herein, we critically evaluate recent findings in the field which illustrate that drebrin E works together with two other actin-binding proteins, namely Arp3 (actin-related protein 3, a component of the Arp2/3 complex that simultaneously controls actin nucleation for polymerization and branching of actin filaments) and Eps8 (epidermal growth factor receptor pathway substrate 8 that controls capping of the barbed ends of actin filaments, as well as actin filament bundling) to regulate the homeostasis of F-actin filament bundles at the ectoplasmic specialization (ES), a testis-specific atypical adherens junction (AJ) in the seminiferous epithelium. This is mediated by the strict temporal and spatial expression of these three actin-binding proteins at the apical and basal ES at the Sertoli cell-spermatid (step 8-19) and Sertoli-Sertoli cell interface, respectively, during the seminiferous epithelial cycle of spermatogenesis. In this Commentary, we put forth a possible model by which drebrin E may be acting as a platform upon which proteins (e.g., Arp3) that are needed to alter the conformation of actin filament bundles at the ES can be recruited to the site, thus facilitating changes in cell shape and cell position in the epithelium during spermiogenesis and spermiation. In short, drebrin E may be acting as a "logistic" distribution center to manage different regulatory proteins at the apical ES, thereby regulating the dynamics of actin filament bundles and modulating the plasticity of the apical ES. This would allow adhesion to be altered continuously throughout the epithelial cycle to accommodate spermatid movement in the seminiferous epithelium during spermiogenesis and spermiation. We also describe a hypothetical model, upon which functional studies can be designed in the future.
Collapse
Affiliation(s)
- C Yan Cheng
- Center for Biomedical Research; The Population Council; New York, NY USA
| | | |
Collapse
|
271
|
Liu Y, Medda R, Liu Z, Galior K, Yehl K, Spatz JP, Cavalcanti-Adam E, Salaita K. Nanoparticle tension probes patterned at the nanoscale: impact of integrin clustering on force transmission. NANO LETTERS 2014; 14:5539-46. [PMID: 25238229 PMCID: PMC4189618 DOI: 10.1021/nl501912g] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 08/29/2014] [Indexed: 05/06/2023]
Abstract
Herein we aimed to understand how nanoscale clustering of RGD ligands alters the mechano-regulation of their integrin receptors. We combined molecular tension fluorescence microscopy with block copolymer micelle nanolithography to fabricate substrates with arrays of precisely spaced probes that can generate a 10-fold fluorescence response to pN-forces. We found that the mechanism of sensing ligand spacing is force-mediated. This strategy is broadly applicable to investigating receptor clustering and its role in mechanotransduction pathways.
Collapse
Affiliation(s)
- Yang Liu
- Department
of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Rebecca Medda
- Department
of Biophysical Chemistry, Institute of Physical Chemistry, Ruprecht-Karls-University, INF 253, 69120 Heidelberg, Germany
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Stuttgart, Germany
| | - Zheng Liu
- Department
of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Kornelia Galior
- Department
of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Kevin Yehl
- Department
of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Joachim P. Spatz
- Department
of Biophysical Chemistry, Institute of Physical Chemistry, Ruprecht-Karls-University, INF 253, 69120 Heidelberg, Germany
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Stuttgart, Germany
| | - Elisabetta
Ada Cavalcanti-Adam
- Department
of Biophysical Chemistry, Institute of Physical Chemistry, Ruprecht-Karls-University, INF 253, 69120 Heidelberg, Germany
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Stuttgart, Germany
| | - Khalid Salaita
- Department
of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| |
Collapse
|
272
|
Bruntz RC, Lindsley CW, Brown HA. Phospholipase D signaling pathways and phosphatidic acid as therapeutic targets in cancer. Pharmacol Rev 2014; 66:1033-79. [PMID: 25244928 PMCID: PMC4180337 DOI: 10.1124/pr.114.009217] [Citation(s) in RCA: 184] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Phospholipase D is a ubiquitous class of enzymes that generates phosphatidic acid as an intracellular signaling species. The phospholipase D superfamily plays a central role in a variety of functions in prokaryotes, viruses, yeast, fungi, plants, and eukaryotic species. In mammalian cells, the pathways modulating catalytic activity involve a variety of cellular signaling components, including G protein-coupled receptors, receptor tyrosine kinases, polyphosphatidylinositol lipids, Ras/Rho/ADP-ribosylation factor GTPases, and conventional isoforms of protein kinase C, among others. Recent findings have shown that phosphatidic acid generated by phospholipase D plays roles in numerous essential cellular functions, such as vesicular trafficking, exocytosis, autophagy, regulation of cellular metabolism, and tumorigenesis. Many of these cellular events are modulated by the actions of phosphatidic acid, and identification of two targets (mammalian target of rapamycin and Akt kinase) has especially highlighted a role for phospholipase D in the regulation of cellular metabolism. Phospholipase D is a regulator of intercellular signaling and metabolic pathways, particularly in cells that are under stress conditions. This review provides a comprehensive overview of the regulation of phospholipase D activity and its modulation of cellular signaling pathways and functions.
Collapse
Affiliation(s)
- Ronald C Bruntz
- Department of Pharmacology (R.C.B., C.W.L., H.A.B.) and Vanderbilt Center for Neuroscience Drug Discovery (C.W.L.), Vanderbilt University Medical Center; Department of Chemistry, Vanderbilt Institute of Chemical Biology (C.W.L., H.A.B.); Vanderbilt Specialized Chemistry for Accelerated Probe Development (C.W.L.); and Department of Biochemistry, Vanderbilt-Ingram Cancer Center (H.A.B.), Vanderbilt University, Nashville, Tennessee
| | - Craig W Lindsley
- Department of Pharmacology (R.C.B., C.W.L., H.A.B.) and Vanderbilt Center for Neuroscience Drug Discovery (C.W.L.), Vanderbilt University Medical Center; Department of Chemistry, Vanderbilt Institute of Chemical Biology (C.W.L., H.A.B.); Vanderbilt Specialized Chemistry for Accelerated Probe Development (C.W.L.); and Department of Biochemistry, Vanderbilt-Ingram Cancer Center (H.A.B.), Vanderbilt University, Nashville, Tennessee
| | - H Alex Brown
- Department of Pharmacology (R.C.B., C.W.L., H.A.B.) and Vanderbilt Center for Neuroscience Drug Discovery (C.W.L.), Vanderbilt University Medical Center; Department of Chemistry, Vanderbilt Institute of Chemical Biology (C.W.L., H.A.B.); Vanderbilt Specialized Chemistry for Accelerated Probe Development (C.W.L.); and Department of Biochemistry, Vanderbilt-Ingram Cancer Center (H.A.B.), Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
273
|
Huda S, Pilans D, Makurath M, Hermans T, Kandere-Grzybowska K, Grzybowski BA. Microfabricated Systems and Assays for Studying the Cytoskeletal Organization, Micromechanics, and Motility Patterns of Cancerous Cells. ADVANCED MATERIALS INTERFACES 2014; 1:1400158. [PMID: 26900544 PMCID: PMC4757490 DOI: 10.1002/admi.201400158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Cell motions are driven by coordinated actions of the intracellular cytoskeleton - actin, microtubules (MTs) and substrate/focal adhesions (FAs). This coordination is altered in metastatic cancer cells resulting in deregulated and increased cellular motility. Microfabrication tools, including photolithography, micromolding, microcontact printing, wet stamping and microfluidic devices have emerged as a powerful set of experimental tools with which to probe and define the differences in cytoskeleton organization/dynamics and cell motility patterns in non-metastatic and metastatic cancer cells. In this review, we discuss four categories of microfabricated systems: (i) micropatterned substrates for studying of cell motility sub-processes (for example, MT targeting of FAs or cell polarization); (ii) systems for studying cell mechanical properties, (iii) systems for probing overall cell motility patterns within challenging geometric confines relevant to metastasis (for example, linear and ratchet geometries), and (iv) microfluidic devices that incorporate co-cultures of multiple cells types and chemical gradients to mimic in vivo intravasation/extravasation steps of metastasis. Together, these systems allow for creating controlled microenvironments that not only mimic complex soft tissues, but are also compatible with live cell high-resolution imaging and quantitative analysis of single cell behavior.
Collapse
Affiliation(s)
- Sabil Huda
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA
| | - Didzis Pilans
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA
| | - Monika Makurath
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA
| | - Thomas Hermans
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA
| | - Kristiana Kandere-Grzybowska
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA
| | - Bartosz A Grzybowski
- Department of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA; Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL, USA
| |
Collapse
|
274
|
Yagi H, Nagano T, Xie MJ, Ikeda H, Kuroda K, Komada M, Iguchi T, Tariqur RM, Morikubo S, Noguchi K, Murase K, Okabe M, Sato M. Filamin A-interacting protein (FILIP) is a region-specific modulator of myosin 2b and controls spine morphology and NMDA receptor accumulation. Sci Rep 2014; 4:6353. [PMID: 25220605 PMCID: PMC4163676 DOI: 10.1038/srep06353] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 08/26/2014] [Indexed: 12/14/2022] Open
Abstract
Learning and memory depend on morphological and functional changes to neural spines. Non-muscle myosin 2b regulates actin dynamics downstream of long-term potentiation induction. However, the mechanism by which myosin 2b is regulated in the spine has not been fully elucidated. Here, we show that filamin A-interacting protein (FILIP) is involved in the control of neural spine morphology and is limitedly expressed in the brain. FILIP bound near the ATPase domain of non-muscle myosin heavy chain IIb, an essential component of myosin 2b, and modified the function of myosin 2b by interfering with its actin-binding activity. In addition, FILIP altered the subcellular distribution of myosin 2b in spines. Moreover, subunits of the NMDA receptor were differently distributed in FILIP-expressing neurons, and excitation propagation was altered in FILIP-knockout mice. These results indicate that FILIP is a novel, region-specific modulator of myosin 2b.
Collapse
Affiliation(s)
- Hideshi Yagi
- 1] Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan [2] Research and Education Program for Life Science, University of Fukui, Fukui 910-8507, Japan [3] Department of Anatomy and Neuroscience, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - Takashi Nagano
- Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Min-Jue Xie
- 1] Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan [2] Research and Education Program for Life Science, University of Fukui, Fukui 910-8507, Japan
| | - Hiroshi Ikeda
- 1] Research and Education Program for Life Science, University of Fukui, Fukui 910-8507, Japan [2] Department of Human and Artificial Intelligence Systems, Faculty of Engineering, University of Fukui, Fukui 910-8507, Japan
| | - Kazuki Kuroda
- 1] Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan [2] Research and Education Program for Life Science, University of Fukui, Fukui 910-8507, Japan
| | - Munekazu Komada
- 1] Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan [2] Research and Education Program for Life Science, University of Fukui, Fukui 910-8507, Japan
| | - Tokuichi Iguchi
- 1] Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan [2] Research and Education Program for Life Science, University of Fukui, Fukui 910-8507, Japan [3] Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Rahman M Tariqur
- Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Soichi Morikubo
- 1] Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan [2] Division of Ophthalmology, Department of Sensory and Locomotor Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Koichi Noguchi
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - Kazuyuki Murase
- 1] Research and Education Program for Life Science, University of Fukui, Fukui 910-8507, Japan [2] Department of Human and Artificial Intelligence Systems, Faculty of Engineering, University of Fukui, Fukui 910-8507, Japan
| | - Masaru Okabe
- Department of Experimental Genome Research, Genome Information Research Center, Osaka University, Osaka 565-0871, Japan
| | - Makoto Sato
- 1] Division of Cell Biology and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan [2] Research and Education Program for Life Science, University of Fukui, Fukui 910-8507, Japan [3] Research Center for Child Mental Development, University of Fukui, Fukui 910-1193, Japan [4] United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Osaka 565-0871, Japan [5] Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
275
|
Lin SP, Huang SY, Chen SF, Vinzons LU, Ciou JY, Wong PJ. Investigation of the interfacial effects of small chemical-modified TiO2 nanotubes on 3T3 fibroblast responses. ACS APPLIED MATERIALS & INTERFACES 2014; 6:12071-12082. [PMID: 25012464 DOI: 10.1021/am503323y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
In order to gain insight into how interfacial effects influence cell responses, chemically modified anodized TiO2 nanotubes (ATNs) were used to simultaneously investigate the effects of nanoscale substrate structure and angstrom-scale chemicals on cell morphological change and cell growth. Two small chemicals were used to modify the ATNs, namely, 3-aminopropyltrimethoxysilane (APTMS) and 3-mercaptopropyltrimethoxysilane (MPTMS), resulting in APTMS-modified ATNs (APTMS-ATNs) and MPTMS-modified ATNs (MPTMS-ATNs), respectively. In our in vitro observation of NIH/3T3 fibroblasts, cells thrived on both unmodified and modified ATNs. Quantitative analyses of cell numbers exhibited that APTMS-ATNs effectively facilitated cell proliferation and directed cell orientation owing to full cell-substrate contact caused by positively charged amino groups (-NH3(+)) on the surface. In addition, scanning electron microscopy and fluorescence images showed different cell morphologies on APTMS-ATNs and MPTMS-ATNs. APTMS-ATNs resulted in flat spreading of fibroblasts, while MPTMS-ATNs resulted in fibroblasts with a three-dimensional solid shape and clear contours. The results indicate that the synergistic effects of nanotube surface topology and small chemical modification and, to a lesser extent, surface hydrophilicity, alter the interfacial interactions between cells and substrates, significantly affecting cell morphology, attachment, and growth. Using ATNs with different interfacial effects from various small chemicals, orientation of cells into various patterns can be achieved and investigation of cell fates, such as proliferation or stem cell differentiation, can be performed for future advanced medical or biological applications.
Collapse
Affiliation(s)
- Shu-Ping Lin
- Graduate Institute of Biomedical Engineering, §Bachelor Program of Biotechnology, National Chung Hsing University , 250 Kuo-Kuang Road, Taichung, 40227 Taiwan
| | | | | | | | | | | |
Collapse
|
276
|
Tripathi BK, Lowy DR, Zelenka PS. The Cdk5 activator P39 specifically links muskelin to myosin II and regulates stress fiber formation and actin organization in lens. Exp Cell Res 2014; 330:186-98. [PMID: 25128817 DOI: 10.1016/j.yexcr.2014.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 07/31/2014] [Accepted: 08/02/2014] [Indexed: 10/24/2022]
Abstract
Cyclin dependent kinase 5 (Cdk5), a proline-directed serine/threonine kinase, requires p39 for its enzymatic activity, and is implicated in cytoskeletal organization and contraction in numerous cell types. The C-terminus of p39 binds muskelin, a multi-domain scaffolding protein known to affect cytoskeletal organization, but the mechanisms by which muskelin affects cytoskeletal organization remain unclear. The present study sought to determine whether p39 might serve as an adaptor protein that links muskelin to stress fibers and to investigate the possible biological relevance of such an interaction. Double immunoprecipitation showed that muskelin, p39, and myosin II are components of a single intracellular complex, and suppressing p39 abrogated the interaction between muskelin and the myosin subunits, demonstrating that p39 is required to link muskelin to myosin II. Muskelin is colocalized with myosin regulatory light chain (MRLC) and on stress fibers. The suppression of muskelin reduced Rho-GTP, MRLC phosphorylation, disrupted stress fiber organization, and promoted cell migration, all of which closely mimic the effect of Cdk5 inhibition. Moreover, suppressing muskelin and inhibiting Cdk5 together have no additional effect, indicating that muskelin plays an important role in Cdk5-dependent signaling. p39 is necessary and sufficient for Cdk5-dependent regulation of MRLC phosphorylation, as suppression of p39, but not p35, reduces MRLC phosphorylation. Together, these results demonstrate that p39 specifically links muskelin to myosin II and consequently, to stress fibers and reveal a novel role for muskelin in regulating myosin phosphorylation and cytoskeletal organization.
Collapse
Affiliation(s)
- Brajendra K Tripathi
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Building 37, Room 4112, Bethesda, MD 20892, USA; Laboratory of Molecular and Developmental Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Douglas R Lowy
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Building 37, Room 4112, Bethesda, MD 20892, USA
| | - Peggy S Zelenka
- Laboratory of Molecular and Developmental Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
277
|
Siani A, Tirelli N. Myofibroblast differentiation: main features, biomedical relevance, and the role of reactive oxygen species. Antioxid Redox Signal 2014; 21:768-85. [PMID: 24279926 DOI: 10.1089/ars.2013.5724] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Myofibroblasts are prototypical fibrotic cells, which are involved in a number of more or less pathological conditions, from foreign body reactions to scarring, from liver, kidney, or lung fibrosis to neoplastic phenomena. The differentiation of precursor cells (not only of fibroblastic nature) is characterized by a complex interplay between soluble factors (growth factors such as transforming growth factor β1, reactive oxygen species [ROS]) and material properties (matrix stiffness). RECENT ADVANCES The last 15 years have seen very significant advances in the identification of appropriate differentiation markers, in the understanding of the differentiation mechanism, and above all, the involvement of ROS as causative and persistence factors. CRITICAL ISSUES The specific mechanisms of action of ROS remain largely unknown, although evidence suggests that both intracellular and extracellular phenomena play a role. FUTURE DIRECTIONS Approaches based on antioxidant (ROS-scavenging) principles and on the potentiation of nitric oxide signaling hold much promise in view of a pharmacological therapy of fibrotic phenomena. However, how to make the active principles available at the target sites is yet a largely neglected issue.
Collapse
Affiliation(s)
- Alessandro Siani
- 1 School of Pharmacy and Pharmaceutical Sciences, University of Manchester , Manchester, United Kingdom
| | | |
Collapse
|
278
|
Zhu L, Yang J, Zhang J, Peng B. A Comparative Study of BioAggregate and ProRoot MTA on Adhesion, Migration, and Attachment of Human Dental Pulp Cells. J Endod 2014; 40:1118-23. [DOI: 10.1016/j.joen.2013.12.028] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 11/24/2013] [Accepted: 12/27/2013] [Indexed: 01/08/2023]
|
279
|
Lal MA, Andersson AC, Katayama K, Xiao Z, Nukui M, Hultenby K, Wernerson A, Tryggvason K. Rhophilin-1 is a key regulator of the podocyte cytoskeleton and is essential for glomerular filtration. J Am Soc Nephrol 2014; 26:647-62. [PMID: 25071083 DOI: 10.1681/asn.2013111195] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Rhophilin-1 is a Rho GTPase-interacting protein, the biologic function of which is largely unknown. Here, we identify and describe the functional role of Rhophilin-1 as a novel podocyte-specific protein of the kidney glomerulus. Rhophilin-1 knockout mice were phenotypically normal at birth but developed albuminuria at about 2 weeks of age. Kidneys from severely albuminuric mice revealed widespread podocyte foot process effacement, thickening of the glomerular basement membrane, and FSGS-like lesions. The absence of any overt changes in the expression of podocyte proteins at the onset of proteinuria suggested that the primary cause of podocyte abnormalities in Rhpn1-null mice was the result of cell-autonomous, Rhophilin-1-dependent signaling events. In culture, Rhophilin-1 was detected at the plasma membrane leading edge of primary podocytes, where it elicited remodeling of the actin cytoskeleton network. This effect of Rhophilin-1 on actin cytoskeleton organization associated with inhibitory effects on Rho-dependent phosphorylation of the myosin regulatory light chain and stress fiber formation. Conversely, phosphorylation of myosin regulatory light chain increased in podocyte foot processes of Rhpn1(-/-) mice, implicating altered actinomyosin contractility in foot process effacement and compromised filtration capacity. Targeted deletion of RhoA in podocytes of Rhophilin-1 knockout mice exacerbated the renal injury. Taken together, our results indicate that Rhophilin-1 is essential for the integrity of the glomerular filtration barrier and that this protein is a key determinant of podocyte cytoskeleton architecture.
Collapse
Affiliation(s)
- Mark A Lal
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics
| | | | - Kan Katayama
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics
| | - Ziejie Xiao
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics
| | - Masatoshi Nukui
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics
| | - Kjell Hultenby
- Department of Laboratory Medicine, Division of Clinical Research Center, and
| | - Annika Wernerson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Karl Tryggvason
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics,
| |
Collapse
|
280
|
Piacentini N, Verkhovsky AB, Gabella C, Meister JJ, Vianay B. Ultra-soft cantilevers and 3-D micro-patterned substrates for contractile bundle tension measurement in living cells. LAB ON A CHIP 2014; 14:2539-2547. [PMID: 24867825 DOI: 10.1039/c4lc00188e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Actin-myosin microfilament bundles or stress-fibers are the principal tension-generating structures in the cell. Their mechanical properties are critical for cell shape, motion, and interaction with other cells and extracellular matrix, but were so far difficult to access in a living cell. Here we propose a micro-fabricated two-component setup for direct tension measurement on a peripheral bundle within an intact cell. We used 3-D substrates made of silicon elastomer to elevate the cell making the filament bundle at its border accessible from the side, and employed an ultra-soft (spring constant 0.78 nN μm(-1)) epoxy-based cantilever for mechanical probing. With this setup we were able for the first time to measure the tension in peripheral actin bundles in living primary fibroblasts spread on a rigid substrate.
Collapse
Affiliation(s)
- Niccolò Piacentini
- Laboratory of Cell Biophysics, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | | | | | | | | |
Collapse
|
281
|
Fife CM, McCarroll JA, Kavallaris M. Movers and shakers: cell cytoskeleton in cancer metastasis. Br J Pharmacol 2014; 171:5507-23. [PMID: 24665826 DOI: 10.1111/bph.12704] [Citation(s) in RCA: 397] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 03/14/2014] [Accepted: 03/18/2014] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Metastasis is responsible for the greatest number of cancer deaths. Metastatic disease, or the movement of cancer cells from one site to another, is a complex process requiring dramatic remodelling of the cell cytoskeleton. The various components of the cytoskeleton, actin (microfilaments), microtubules (MTs) and intermediate filaments, are highly integrated and their functions are well orchestrated in normal cells. In contrast, mutations and abnormal expression of cytoskeletal and cytoskeletal-associated proteins play an important role in the ability of cancer cells to resist chemotherapy and metastasize. Studies on the role of actin and its interacting partners have highlighted key signalling pathways, such as the Rho GTPases, and downstream effector proteins that, through the cytoskeleton, mediate tumour cell migration, invasion and metastasis. An emerging role for MTs in tumour cell metastasis is being unravelled and there is increasing interest in the crosstalk between key MT interacting proteins and the actin cytoskeleton, which may provide novel treatment avenues for metastatic disease. Improved understanding of how the cytoskeleton and its interacting partners influence tumour cell migration and metastasis has led to the development of novel therapeutics against aggressive and metastatic disease. LINKED ARTICLES This article is part of a themed section on Cytoskeleton, Extracellular Matrix, Cell Migration, Wound Healing and Related Topics. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2014.171.issue-24.
Collapse
Affiliation(s)
- C M Fife
- Tumour Biology and Targeting Program, Children's Cancer Institute Australia Lowy Cancer Research Centre, UNSW Australia, Randwick, NSW, Australia; Australian Centre for NanoMedicine, UNSW Australia, Sydney, NSW, Australia
| | | | | |
Collapse
|
282
|
Koride S, He L, Xiong LP, Lan G, Montell DJ, Sun SX. Mechanochemical regulation of oscillatory follicle cell dynamics in the developing Drosophila egg chamber. Mol Biol Cell 2014; 25:3709-16. [PMID: 24943847 PMCID: PMC4230628 DOI: 10.1091/mbc.e14-04-0875] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In the epithelium of Drosophila during tissue elongation, contractile forces in follicle cells can oscillate. These oscillations correlate with increasing tension in the epithelium from egg chamber growth. A mathematical model is proposed to explain the observed oscillations, together with a mechanism of active regulation of cellular contractile forces. During tissue elongation from stage 9 to stage 10 in Drosophila oogenesis, the egg chamber increases in length by ∼1.7-fold while increasing in volume by eightfold. During these stages, spontaneous oscillations in the contraction of cell basal surfaces develop in a subset of follicle cells. This patterned activity is required for elongation of the egg chamber; however, the mechanisms generating the spatiotemporal pattern have been unclear. Here we use a combination of quantitative modeling and experimental perturbation to show that mechanochemical interactions are sufficient to generate oscillations of myosin contractile activity in the observed spatiotemporal pattern. We propose that follicle cells in the epithelial layer contract against pressure in the expanding egg chamber. As tension in the epithelial layer increases, Rho kinase signaling activates myosin assembly and contraction. The activation process is cooperative, leading to a limit cycle in the myosin dynamics. Our model produces asynchronous oscillations in follicle cell area and myosin content, consistent with experimental observations. In addition, we test the prediction that removal of the basal lamina will increase the average oscillation period. The model demonstrates that in principle, mechanochemical interactions are sufficient to drive patterning and morphogenesis, independent of patterned gene expression.
Collapse
Affiliation(s)
- Sarita Koride
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Li He
- Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, Boston, MA 02115
| | - Li-Ping Xiong
- Department of Physics, George Washington University, Washington, DC 20052
| | - Ganhui Lan
- Department of Physics, George Washington University, Washington, DC 20052
| | - Denise J Montell
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106
| | - Sean X Sun
- Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218 Department of Mechanical Engineering, Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218
| |
Collapse
|
283
|
Abstract
Nephrotic syndrome (NS) is characterized by heavy proteinuria, hypoalbuminemia, and edema. The underlying causes of NS are diverse and are tied to inheritable and environmental factors. A common diagnostic marker for NS is effacement of podocyte foot processes. The formation and maintenance of foot processes are under the control of many signalling molecules including Rho-GTPases. Our knowledge of Rho-GTPases is based largely on the functions of three prototypic members: RhoA, Rac1, and Cdc42. In the event of podocyte injury, the rearrangement to the actin cytoskeleton is orchestrated largely by this family of proteins. The importance of maintaining proper actin dynamics in podocytes has led to much investigation as to how Rho-GTPases and their regulatory molecules form and maintain foot processes as a critical component of the kidney’s filtration barrier. Modern sequencing techniques have allowed for the identification of novel disease causing mutations in genes such as ARHGDIA, encoding Rho-GDIα. Continued use of whole exome sequencing has the potential to lead to the identification of new mutations in genes encoding Rho-GTPases or their regulatory proteins. Expanding our knowledge of the dynamic regulation of the actin network by Rho-GTPases in podocytes will pave the way for effective therapeutic options for NS patients.
Collapse
|
284
|
Horne-Badovinac S. The Drosophila egg chamber-a new spin on how tissues elongate. Integr Comp Biol 2014; 54:667-76. [PMID: 24920751 DOI: 10.1093/icb/icu067] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
During development, tissues undergo complex cellular rearrangements and changes in shape that produce a diversity of body plans and the functional organs therein. The Drosophila egg chamber has emerged as an exciting and highly tractable model in which to investigate novel mechanisms driving the elongation of tissues. Egg chambers are multicellular assemblies within flies' ovaries that will each give rise to a single egg. Although initially spherical, these simple organ-like structures lengthen as they grow. This transformation depends on an unusual form of planar polarity in the egg chamber's outer epithelial layer, in which arrays of linear actin bundles and fibril-like structures in the basement membrane both align perpendicular to the axis of elongation. The resulting circumferential arrangement of structural molecules is then thought to act as a "molecular corset" that directionally biases growth of the egg chamber. I will explore four fundamental questions about this system: (1) How is the circumferential pattern generated in the follicular epithelium? (2) What is the physical nature of the corset? (3) How does a corset-type mechanism lead to the cellular rearrangements necessary for the elongation of tissues? and (4) To what extent are the cellular mechanisms controlling egg chamber elongation conserved in other systems? For each topic, I will present insights gleaned from the recent literature and highlight fertile areas for future investigation.
Collapse
Affiliation(s)
- Sally Horne-Badovinac
- Department of Molecular Genetics and Cell Biology, Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, 920 East 58th Street, Chicago, IL 60637, USA
| |
Collapse
|
285
|
Smith MA, Hoffman LM, Beckerle MC. LIM proteins in actin cytoskeleton mechanoresponse. Trends Cell Biol 2014; 24:575-83. [PMID: 24933506 DOI: 10.1016/j.tcb.2014.04.009] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 04/23/2014] [Accepted: 04/30/2014] [Indexed: 01/21/2023]
Abstract
The actin cytoskeleton assembles into branched networks or bundles to generate mechanical force for critical cellular processes such as establishment of polarity, adhesion, and migration. Stress fibers (SFs) are contractile actomyosin structures that physically couple to the extracellular matrix through integrin-based focal adhesions (FAs), thereby transmitting force into and across the cell. Recently, LIN-11, Isl1, and MEC-3 (LIM) domain proteins have been implicated in mediating this cytoskeletal mechanotransduction. Among the more well-studied LIM domain adapter proteins is zyxin, a dynamic component of both FAs and SFs. Here we discuss recent research detailing the mechanisms by which SFs adjust their structure and composition to balance mechanical forces and suggest ways that zyxin and other LIM domain proteins mediate mechanoresponse.
Collapse
Affiliation(s)
- M A Smith
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - L M Hoffman
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - M C Beckerle
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
286
|
Kutscheidt S, Zhu R, Antoku S, Luxton GWG, Stagljar I, Fackler OT, Gundersen GG. FHOD1 interaction with nesprin-2G mediates TAN line formation and nuclear movement. Nat Cell Biol 2014; 16:708-15. [PMID: 24880667 PMCID: PMC4113092 DOI: 10.1038/ncb2981] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/30/2014] [Indexed: 12/26/2022]
Abstract
Active positioning of the nucleus is integral to division, migration, and differentiation of mammalian cells1. Fibroblasts polarizing for migration orient their centrosomes by actin-dependent nuclear movement2. This nuclear movement depends on nesprin-2 giant (N2G), a large, actin-binding outer nuclear membrane component of transmembrane actin-associated (TAN) lines that couple nuclei to moving actin cables3. Here, we identify the diaphanous formin FHOD1 as an interaction partner of N2G. Silencing FHOD1 expression or expression of fragments containing binding sites of N2G or FHOD1 disrupted nuclear movement and centrosome orientation in polarizing fibroblasts. Unexpectedly, silencing of FHOD1 expression did not affect the formation or rearward flow of dorsal actin cables required for nuclear positioning. Rather, N2G-FHOD1 interaction provided a second connection to actin cables essential for TAN line formation and thus nuclear movement. These results reveal a unique function for a formin in coupling an organelle to actin filaments for translocation and suggest that TAN lines require multi-point attachments to actin cables to resist the large forces necessary to move the nucleus.
Collapse
Affiliation(s)
- Stefan Kutscheidt
- 1] Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, Heidelberg, INF 324, 69120 Heidelberg, Germany [2]
| | - Ruijun Zhu
- 1] Department of Pathology and Cell Biology, Columbia University, New York, New York 10032, USA [2]
| | - Susumu Antoku
- 1] Department of Pathology and Cell Biology, Columbia University, New York, New York 10032, USA [2]
| | - G W Gant Luxton
- 1] Department of Pathology and Cell Biology, Columbia University, New York, New York 10032, USA [2]
| | - Igor Stagljar
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Oliver T Fackler
- Department of Infectious Diseases, Integrative Virology, University Hospital Heidelberg, Heidelberg, INF 324, 69120 Heidelberg, Germany
| | - Gregg G Gundersen
- Department of Pathology and Cell Biology, Columbia University, New York, New York 10032, USA
| |
Collapse
|
287
|
Muenzner JK, Biersack B, Kalie H, Andronache IC, Kaps L, Schuppan D, Sasse F, Schobert R. Gold(I) biscarbene complexes derived from vascular-disrupting combretastatin A-4 address different targets and show antimetastatic potential. ChemMedChem 2014; 9:1195-1204. [PMID: 24648184 DOI: 10.1002/cmdc.201400049] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Indexed: 11/08/2022]
Abstract
Gold N-heterocyclic carbene (NHC) complexes are an emerging class of anticancer drugs. We present a series of gold(I) biscarbene complexes with NHC ligands derived from the plant metabolite combretastatin A-4 (CA-4) that retain its vascular-disrupting effect, yet address different cellular and protein targets. Unlike CA-4, these complexes did not interfere with tubulin, but with the actin cytoskeleton of endothelial and cancer cells. For the highly metastatic 518A2 melanoma cell line this effect was accompanied by a marked accumulation of cells in the G1 phase of the cell cycle and a suppression of active prometastatic matrix metalloproteinase-2. Despite these mechanistic differences the complexes were as strongly antivascular as CA-4 both in vitro in tube formation assays with human umbilical vein endothelial cells, and in vivo as to blood vessel disruption in the chorioallantoic membrane of chicken eggs. The antiproliferative effect of the new gold biscarbene complexes in a panel of six human cancer cell lines was impressive, with low sub-micromolar IC50 values (72 h) even against CA-4-refractory HT-29 colon and multidrug-resistant MCF-7 breast carcinoma cells. In preliminary studies with a mouse melanoma xenograft model the complexes led to significant decreases in tumor volume while being very well tolerated.
Collapse
Affiliation(s)
- Julienne K Muenzner
- Department of Organic Chemistry, University Bayreuth, Universitätsstraße 30, 95440 Bayreuth (Germany)
| | | | | | | | | | | | | | | |
Collapse
|
288
|
Cadinu D, Hooda J, Alam MM, Balamurugan P, Henke RM, Zhang L. Comparative proteomic analysis reveals characteristic molecular changes accompanying the transformation of nonmalignant to cancer lung cells. EUPA OPEN PROTEOMICS 2014. [DOI: 10.1016/j.euprot.2014.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
289
|
Contact guidance of smooth muscle cells is associated with tension-mediated adhesion maturation. Exp Cell Res 2014; 327:1-11. [PMID: 24825188 DOI: 10.1016/j.yexcr.2014.05.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 04/23/2014] [Accepted: 05/03/2014] [Indexed: 12/24/2022]
Abstract
Contact guidance is a cellular phenomenon observed during wound healing and developmental patterning, in which adherent cells align in the same direction due to physical cues. Despite numerous studies, the molecular mechanism underlying the consistent cell orientation is poorly understood. Here we fabricated microgrooves with a pitch of submicrons to study contact guidance of smooth muscle cells. We show that both integrin-based cell-substrate adhesions and cellular tension are necessary to achieve contact guidance along microgrooves. We further show through analyses on paxillin that cell-substrate adhesions are more prone to become mature when they run along microgrooves than align at an angle to the direction of microgrooves. Because cellular tension promotes the maturation of cell-substrate adhesions, we propose that the adhesions aligning across microgrooves are not physically efficient for bearing cellular tension compared to those aligning along microgrooves. Thus, the proposed model describes a mechanism of contact guidance that cells would finally align preferentially along microgrooves because cellular tensions are more easily borne within the direction, and the direction of resulting mature adhesions determines the direction of the whole cells.
Collapse
|
290
|
Chan E, Saito A, Honda T, Di Guglielmo GM. The acetylenic tricyclic bis(cyano enone), TBE-31 inhibits non-small cell lung cancer cell migration through direct binding with actin. Cancer Prev Res (Phila) 2014; 7:727-37. [PMID: 24806663 DOI: 10.1158/1940-6207.capr-13-0403] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The migratory and invasive potential of the epithelial-derived tumor cells depends on epithelial-to-mesenchymal transition (EMT) as well as the reorganization of the cell cytoskeleton. Here, we show that the tricyclic compound acetylenic tricyclic bis(cyano enone), TBE-31, directly binds to actin and inhibits linear and branched actin polymerization in vitro. Furthermore, we observed that TBE-31 inhibits stress fiber formation in fibroblasts as well as in non-small cell lung cancer cells during TGFβ-dependent EMT. Interestingly, TBE-31 does not interfere with TGFβ-dependent signaling or changes in E-cadherin and N-cadherin protein levels during EMT. Finally, we observed that TBE-31 inhibits fibroblast and non-small cell lung tumor cell migration with an IC50 of 1.0 and 2.5 μmol/L, respectively. Taken together, our results suggest that TBE-31 targets linear actin polymerization to alter cell morphology and inhibit cell migration.
Collapse
Affiliation(s)
- Eddie Chan
- Authors' Affiliations: Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Akira Saito
- Institute of Chemical Biology and Drug Discovery; and
| | - Tadashi Honda
- Institute of Chemical Biology and Drug Discovery; and Department of Chemistry, Stony Brook University, Stony Brook, New York
| | - Gianni M Di Guglielmo
- Authors' Affiliations: Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada;
| |
Collapse
|
291
|
Maraldi M, Garikipati K. The mechanochemistry of cytoskeletal force generation. Biomech Model Mechanobiol 2014; 14:59-72. [PMID: 24796414 DOI: 10.1007/s10237-014-0588-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 04/21/2014] [Indexed: 01/09/2023]
Abstract
In this communication, we propose a model to study the non-equilibrium process by which actin stress fibers develop force in contractile cells. The emphasis here is on the non-equilibrium thermodynamics, which is necessary to address the mechanics as well as the chemistry of dynamic cell contractility. In this setting, we are able to develop a framework that relates (a) the dynamics of force generation within the cell and (b) the cell's response to external stimuli to the chemical processes occurring within the cell, as well as to the mechanics of linkage between the stress fibers, focal adhesions and extracellular matrix.
Collapse
Affiliation(s)
- Mirko Maraldi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA,
| | | |
Collapse
|
292
|
Renga B, Francisci D, Schiaroli E, Carino A, Cipriani S, D'Amore C, Sidoni A, Sordo RD, Ferri I, Lucattelli M, Lunghi B, Baldelli F, Fiorucci S. The HIV matrix protein p17 promotes the activation of human hepatic stellate cells through interactions with CXCR2 and Syndecan-2. PLoS One 2014; 9:e94798. [PMID: 24736615 PMCID: PMC3988079 DOI: 10.1371/journal.pone.0094798] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/19/2014] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The human immunodeficiency virus type 1 (HIV-1) p17 is a matrix protein involved in virus life's cycle. CXCR2 and Syndecan-2, the two major coreceptors for the p17 protein, are expressed in hepatic stellate cells (HSCs), a key cell type involved in matrix deposition in liver fibrotic disorders. AIM In this report we have investigated the in vitro impact of p17 on HSCs transdifferentiation and function and underlying signaling pathways involved in these processes. METHODS LX-2 cells, a human HSC line, and primary HSC were challenged with p17 and expressions of fibrogenic markers and of p17 receptors were assessed by qRT-PCR and Western blot. Downstream intracellular signaling pathways were evaluated with qRT-PCR and Western blot as well as after pre-treatment with specific pathway inhibitors. RESULTS Exposure of LX2 cells to p17 increases their contractile force, reshapes the cytoskeleton fibers and upregulates the expression of transdifferentiation markers including αSMA, COL1α1 and endothelin-1 through the activation of Jak/STAT and Rho signaling pathways. These effects are lost in HSCs pre-incubated with a serum from HIV positive person who underwent a vaccination with a p17 peptide. Confocal laser microscopy studies demonstrates that CXCR2 and syndecan-2 co-associate at the plasma membrane after exposure to p17. Immunostaining of HIV/HCV liver biopsies from co-infected patients reveals that the progression of liver fibrosis correlates with a reduced expression of CXCR2. CONCLUSIONS The HIV matrix protein p17 is pro-fibrogenic through its interactions both with CXCR2 and syndecan-2 on activated HSCs.
Collapse
Affiliation(s)
- Barbara Renga
- Department of Experimental and Clinical Medicine, University of Perugia, Perugia, Italy
| | - Daniela Francisci
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Elisabetta Schiaroli
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Adriana Carino
- Department of Experimental and Clinical Medicine, University of Perugia, Perugia, Italy
| | - Sabrina Cipriani
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Claudio D'Amore
- Department of Experimental and Clinical Medicine, University of Perugia, Perugia, Italy
| | - Angelo Sidoni
- Department of Experimental Medicine and Biochemical Sciences, Section of Anatomic Pathology and Histology, University of Perugia, Perugia, Italy
| | - Rachele Del Sordo
- Department of Experimental Medicine and Biochemical Sciences, Section of Anatomic Pathology and Histology, University of Perugia, Perugia, Italy
| | - Ivana Ferri
- Department of Experimental Medicine and Biochemical Sciences, Section of Anatomic Pathology and Histology, University of Perugia, Perugia, Italy
| | | | | | - Franco Baldelli
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Stefano Fiorucci
- Department of Experimental and Clinical Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
293
|
Pritchard RH, Huang YYS, Terentjev EM. Mechanics of biological networks: from the cell cytoskeleton to connective tissue. SOFT MATTER 2014; 10:1864-84. [PMID: 24652375 DOI: 10.1039/c3sm52769g] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
From the cell cytoskeleton to connective tissues, fibrous networks are ubiquitous in metazoan life as the key promoters of mechanical strength, support and integrity. In recent decades, the application of physics to biological systems has made substantial strides in elucidating the striking mechanical phenomena observed in such networks, explaining strain stiffening, power law rheology and cytoskeletal fluidisation - all key to the biological function of individual cells and tissues. In this review we focus on the current progress in the field, with a primer into the basic physics of individual filaments and the networks they form. This is followed by a discussion of biological networks in the context of a broad spread of recent in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Robyn H Pritchard
- Cavendish Laboratory, University of Cambridge, Cambridge, CB3 0HE, UK.
| | | | | |
Collapse
|
294
|
Abstract
The mechanical characteristics of endothelial cells reveal four distinct compartments, namely glycocalyx, cell cortex, cytoplasm and nucleus. There is accumulating evidence that endothelial nanomechanics of these individual compartments control vascular physiology. Depending on protein composition, filament formation and interaction with cross-linker proteins, these four compartments determine endothelial stiffness. Structural organization and mechanical properties directly influence physiological processes such as endothelial barrier function, nitric oxide release and gene expression. This review will focus on endothelial nanomechanics and its impact on vascular function.
Collapse
|
295
|
Chun JN, Kim SY, Park EJ, Kwon EJ, Bae DJ, Kim IS, Kim HK, Park JK, Lee SW, Park HH, So I, Jeon JH. Schisandrin B suppresses TGFβ1-induced stress fiber formation by inhibiting myosin light chain phosphorylation. JOURNAL OF ETHNOPHARMACOLOGY 2014; 152:364-371. [PMID: 24486209 DOI: 10.1016/j.jep.2014.01.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 12/24/2013] [Accepted: 01/21/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Schisandra chinensis fruit extract (SCE) has been used as a traditional oriental medicine for treating vascular diseases. However, the pharmacologic effects and mechanisms of SCE on vascular fibrosis are still largely unknown. Transforming growth factor β1 (TGFβ1)-mediated cellular changes are closely associated with the pathogenesis of vascular fibrotic diseases. Particularly, TGFβ1 induces actin stress fiber formation that is a crucial mechanism underlying vascular smooth muscle cell (VSMC) migration in response to vascular injury. In this study, we investigated the effect of SCE and its active ingredients on TGFβ1-induced stress fiber assembly in A7r5 VSMCs. MATERIALS AND METHODS To investigate pharmacological actions of SCE and its ingredients on TGFβ1-treated VSMCs, we have employed molecular and cell biological technologies, such as confocal microscopy, fluorescence resonance energy transfer, western blotting, and radiometric enzyme analyses. RESULTS We found that SCE inhibited TGFβ1-induced stress fiber formation and cell migration. Schisandrin B (SchB) showed the most prominent effect among the active ingredients of SCE tested. SchB reduced TGFβ1-mediated phosphorylation of myosin light chain, and this effect was independent of RhoA/Rho-associated kinase pathway. Fluorescence resonance energy transfer and radiometric enzyme assays confirmed that SchB inhibited myosin light chain kinase activity. We also showed that SchB decreased TGFβ1-mediated induction of α-smooth muscle actin by inhibiting Smad signaling. CONCLUSIONS The present study demonstrates that SCE and its active ingredient SchB suppressed TGFβ1-induced stress fiber formation at the molecular level. Therefore, our findings may help future investigations to develop multi-targeted therapeutic strategies that attenuate VSMC migration and vascular fibrosis.
Collapse
Affiliation(s)
- Jung Nyeo Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul 110-799, Republic of Korea
| | - Sang-Yeob Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 138-736, Republic of Korea; Department of Medicine, University of Ulsan, College of Medicine, Seoul 138-736, Republic of Korea
| | - Eun-Jung Park
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Eun Jung Kwon
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Dong-Jun Bae
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea
| | - In-San Kim
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea
| | - Hye Kyung Kim
- Department of Urology, Medical School and Institute for Medical Sciences, Chonbuk National University, Jeonju 561-712, Republic of Korea
| | - Jong Kwan Park
- Department of Urology, Medical School and Institute for Medical Sciences, Chonbuk National University, Jeonju 561-712, Republic of Korea
| | - Sung Won Lee
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Republic of Korea
| | - Hyun Ho Park
- Department of Biotechnology, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - Insuk So
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul 110-799, Republic of Korea
| | - Ju-Hong Jeon
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul 110-799, Republic of Korea.
| |
Collapse
|
296
|
Shao Y, Mann JM, Chen W, Fu J. Global architecture of the F-actin cytoskeleton regulates cell shape-dependent endothelial mechanotransduction. Integr Biol (Camb) 2014; 6:300-11. [PMID: 24435061 PMCID: PMC3963173 DOI: 10.1039/c3ib40223a] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Uniaxial stretch is an important biophysical regulator of cell morphology (or shape) and functions of vascular endothelial cells (ECs). However, it is unclear whether and how cell shape can independently regulate EC mechanotransductive properties under uniaxial stretch. Herein, utilizing a novel uniaxial cell-stretching device integrated with micropost force sensors, we reported the first experimental evidence showing cell shape-dependent EC mechanotransduction via cytoskeleton (CSK) contractile forces in response to uniaxial stretch. Combining experiments and theoretical modeling from first principles, we showed that it was the global architecture of the F-actin CSK that instructed the cell shape-dependent EC mechanotransductive process. Furthermore, a cell shape-dependent nature was relayed in EC mechanotransduction via dynamic focal adhesion (FA) assembly. Our results suggested a novel mechanotransductive process in ECs wherein the global architecture of the F-actin CSK, governed by cell shape, controls mechanotransduction via CSK contractile forces and force-dependent FA assembly under uniaxial stretch.
Collapse
Affiliation(s)
- Yue Shao
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA.
| | | | | | | |
Collapse
|
297
|
Fogh BS, Multhaupt HAB, Couchman JR. Protein kinase C, focal adhesions and the regulation of cell migration. J Histochem Cytochem 2014; 62:172-84. [PMID: 24309511 PMCID: PMC3935447 DOI: 10.1369/0022155413517701] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 11/21/2013] [Indexed: 12/18/2022] Open
Abstract
Cell adhesion to extracellular matrix is a complex process involving protrusive activity driven by the actin cytoskeleton, engagement of specific receptors, followed by signaling and cytoskeletal organization. Thereafter, contractile and endocytic/recycling activities may facilitate migration and adhesion turnover. Focal adhesions, or focal contacts, are widespread organelles at the cell-matrix interface. They arise as a result of receptor interactions with matrix ligands, together with clustering. Recent analysis shows that focal adhesions contain a very large number of protein components in their intracellular compartment. Among these are tyrosine kinases, which have received a great deal of attention, whereas the serine/threonine kinase protein kinase C has received much less. Here the status of protein kinase C in focal adhesions and cell migration is reviewed, together with discussion of its roles and potential substrates.
Collapse
Affiliation(s)
- Betina S Fogh
- Department of Biomedical Sciences, University of Copenhagen, Denmark
| | | | | |
Collapse
|
298
|
Blanchoin L, Boujemaa-Paterski R, Sykes C, Plastino J. Actin dynamics, architecture, and mechanics in cell motility. Physiol Rev 2014; 94:235-63. [PMID: 24382887 DOI: 10.1152/physrev.00018.2013] [Citation(s) in RCA: 926] [Impact Index Per Article: 84.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tight coupling between biochemical and mechanical properties of the actin cytoskeleton drives a large range of cellular processes including polarity establishment, morphogenesis, and motility. This is possible because actin filaments are semi-flexible polymers that, in conjunction with the molecular motor myosin, can act as biological active springs or "dashpots" (in laymen's terms, shock absorbers or fluidizers) able to exert or resist against force in a cellular environment. To modulate their mechanical properties, actin filaments can organize into a variety of architectures generating a diversity of cellular organizations including branched or crosslinked networks in the lamellipodium, parallel bundles in filopodia, and antiparallel structures in contractile fibers. In this review we describe the feedback loop between biochemical and mechanical properties of actin organization at the molecular level in vitro, then we integrate this knowledge into our current understanding of cellular actin organization and its physiological roles.
Collapse
|
299
|
Duan X, Chen KL, Zhang Y, Cui XS, Kim NH, Sun SC. ROCK inhibition prevents early mouse embryo development. Histochem Cell Biol 2014; 142:227-33. [PMID: 24562870 DOI: 10.1007/s00418-014-1201-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2014] [Indexed: 12/13/2022]
Abstract
ROCK is a Rho-GTPase effector that is important for actin assembly and is involved in various cellular functions, including cell contraction, migration, motility, and tumor cell invasion. In this study, we investigated ROCK expression and function during early mouse embryo development. Inhibiting ROCK by Y-27632 treatment at the zygote stage resulted in first cleavage failure, and most embryos failed to develop to the 8-cell stage. When adding Y-27632 at the 8-cell stage, embryos failed to undergo compaction and could not develop into blastocysts. In addition, fluorescence staining intensity analysis indicated that actin expression at blastomere membranes was significantly reduced. After ROCK inhibition, two or more nuclei were observed in a cell, which indicated possible cytokinesis failure. Moreover, after ROCK inhibition with Y-27632, the phosphorylation levels of LIMK1/2, a downstream molecule of ROCK, were decreased at blastomere membranes. Thus, our results showed conserved roles for ROCK in this mammalian embryo model and indicated that a ROCK-LIMK1/2-actin pathway might regulate cleavage and blastocyst formation during early mouse embryo development.
Collapse
Affiliation(s)
- Xing Duan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | | | | | | | | | | |
Collapse
|
300
|
Saaber D, Wollenhaupt S, Baumann K, Reichl S. Recent progress in tight junction modulation for improving bioavailability. Expert Opin Drug Discov 2014; 9:367-81. [PMID: 24558958 DOI: 10.1517/17460441.2014.892070] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Currently, there are many novel drugs that belong to class III or IV of the Biopharmaceutics Classification System, showing low bioavailability. Tight junction (TJ) modulation offers an approach to increase bioavailability of pharmaceutical compounds. Furthermore, some diseases are accompanied by disturbed barrier function or TJ dysregulation and thus represent a second application for TJ modulators. AREAS COVERED This review contains a summary of three different TJ modulators: AT1002, PN159 and labradimil. Within this summary, the authors provide a description of their effects on TJs, their adverse effects and their success in clinical trials. Furthermore, the authors present the current understanding of TJ regulation and highlight opportunities to develop new TJ modulators; they also review the problems that might occur. EXPERT OPINION The development of new mechanism-based (MB) TJ modulators is a very promising field of research. MB approaches are expected to have the best future prospects. Further elucidation of signaling pathways and TJ regulation will be necessary for advancing MB TJ modulator research.
Collapse
Affiliation(s)
- Daniel Saaber
- Technische Universität Braunschweig, Institut für Pharmazeutische Technologie , Mendelssohnstr. 1, Braunschweig 38106 , Germany
| | | | | | | |
Collapse
|