201
|
Fibulin-6 regulates pro-fibrotic TGF-β responses in neonatal mouse ventricular cardiac fibroblasts. Sci Rep 2017; 7:42725. [PMID: 28209981 PMCID: PMC5314373 DOI: 10.1038/srep42725] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 01/12/2017] [Indexed: 11/26/2022] Open
Abstract
Fibulin-6, an essential component of extracellular matrix determines the architecture of cellular junctions in tissues undergoing strain. Increased expression and deposition of fibulin-6 facilitates fibroblast migration in response to TGF-β, following myocardial infarction in mouse heart. The underlying mechanism still remains elusive. In conjunction with our previous study, we have now demonstrated that in fibulin-6 knockdown (KD) fibroblasts, not only TGF-β dependent migration, but also stress fiber formation, cellular networking and subsequently fibroblast wound contraction is almost abrogated. SMAD dependent TGF-β pathway shows ~75% decreased translocation of R-SMAD and co-SMAD into the nucleus upon fibulin-6 KD. Consequently, SMAD dependent pro-fibrotic gene expression is considerably down regulated to basal levels both in mRNA and protein. Also, investigating the non-SMAD pathways we observed a constitutive increase in pERK-levels in fibulin-6 KD fibroblast compared to control, but no change was seen in pAKT. Immunoprecipitation studies revealed 60% reduced interaction of TGF-β receptor II and I (TGFRII and I) accompanied by diminished phosphorylation of TGFRI at serin165 in fibulin-6 KD cells. In conclusion, fibulin-6 plays an important role in regulating TGF-β mediated responses, by modulating TGF-β receptor dimerization and activation to further trigger downstream pathways.
Collapse
|
202
|
Weitkunat M, Brasse M, Bausch AR, Schnorrer F. Mechanical tension and spontaneous muscle twitching precede the formation of cross-striated muscle in vivo. Development 2017; 144:1261-1272. [PMID: 28174246 PMCID: PMC5399620 DOI: 10.1242/dev.140723] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 01/28/2017] [Indexed: 02/05/2023]
Abstract
Muscle forces are produced by repeated stereotypical actomyosin units called sarcomeres. Sarcomeres are chained into linear myofibrils spanning the entire muscle fiber. In mammalian body muscles, myofibrils are aligned laterally, resulting in their typical cross-striated morphology. Despite this detailed textbook knowledge about the adult muscle structure, it is still unclear how cross-striated myofibrils are built in vivo. Here, we investigate the morphogenesis of Drosophila abdominal muscles and establish them as an in vivo model for cross-striated muscle development. By performing live imaging, we find that long immature myofibrils lacking a periodic actomyosin pattern are built simultaneously in the entire muscle fiber and then align laterally to give mature cross-striated myofibrils. Interestingly, laser micro-lesion experiments demonstrate that mechanical tension precedes the formation of the immature myofibrils. Moreover, these immature myofibrils do generate spontaneous Ca2+-dependent contractions in vivo, which, when chemically blocked, result in cross-striation defects. Taken together, these results suggest a myofibrillogenesis model in which mechanical tension and spontaneous muscle twitching synchronize the simultaneous self-organization of different sarcomeric protein complexes to build highly regular cross-striated myofibrils spanning the length of large muscle fibers. Summary: In Drosophila, immature myofibrils are built simultaneously across an entire muscle fiber, and then self-organize in a manner dependent on spontaneous contractions and mechanical tension.
Collapse
Affiliation(s)
- Manuela Weitkunat
- Muscle Dynamics Group, Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried 82152, Germany
| | - Martina Brasse
- Lehrstuhl für Biophysik E27, Technische Universität München, James-Franck-Straße 1, Garching 85748, Germany
| | - Andreas R Bausch
- Lehrstuhl für Biophysik E27, Technische Universität München, James-Franck-Straße 1, Garching 85748, Germany
| | - Frank Schnorrer
- Muscle Dynamics Group, Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried 82152, Germany .,Developmental Biology Institute of Marseille (IBDM), CNRS, UMR 7288, Aix-Marseille Université, Case 907, Parc Scientifique de Luminy, Marseille 13288, France
| |
Collapse
|
203
|
Hu X, De Silva TM, Chen J, Faraci FM. Cerebral Vascular Disease and Neurovascular Injury in Ischemic Stroke. Circ Res 2017; 120:449-471. [PMID: 28154097 PMCID: PMC5313039 DOI: 10.1161/circresaha.116.308427] [Citation(s) in RCA: 299] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/13/2016] [Accepted: 10/26/2016] [Indexed: 12/13/2022]
Abstract
The consequences of cerebrovascular disease are among the leading health issues worldwide. Large and small cerebral vessel disease can trigger stroke and contribute to the vascular component of other forms of neurological dysfunction and degeneration. Both forms of vascular disease are driven by diverse risk factors, with hypertension as the leading contributor. Despite the importance of neurovascular disease and subsequent injury after ischemic events, fundamental knowledge in these areas lag behind our current understanding of neuroprotection and vascular biology in general. The goal of this review is to address select key structural and functional changes in the vasculature that promote hypoperfusion and ischemia, while also affecting the extent of injury and effectiveness of therapy. In addition, as damage to the blood-brain barrier is one of the major consequences of ischemia, we discuss cellular and molecular mechanisms underlying ischemia-induced changes in blood-brain barrier integrity and function, including alterations in endothelial cells and the contribution of pericytes, immune cells, and matrix metalloproteinases. Identification of cell types, pathways, and molecules that control vascular changes before and after ischemia may result in novel approaches to slow the progression of cerebrovascular disease and lessen both the frequency and impact of ischemic events.
Collapse
Affiliation(s)
- Xiaoming Hu
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - T. Michael De Silva
- Biomedicine Discovery Institute, Department of Pharmacology, 9 Ancora Imparo Way, Monash University, Clayton, Vic, Australia
| | - Jun Chen
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Frank M. Faraci
- Departments of Internal Medicine and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City Veterans Affairs Healthcare System, Iowa City, IA, USA
| |
Collapse
|
204
|
Center or periphery? Modeling the effects of focal adhesion placement during cell spreading. PLoS One 2017; 12:e0171430. [PMID: 28158263 PMCID: PMC5291721 DOI: 10.1371/journal.pone.0171430] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 01/20/2017] [Indexed: 01/06/2023] Open
Abstract
Focal adhesions are often observed at the cell's periphery. We provide an explanation for this observation using a system-level mathematical model of a cell interacting with a two-dimensional substrate. The model describes the biological cell as a hypoelastic continuum material whose behavior is coupled to a deformable, linear elastic substrate via focal adhesions that are represented by collections of linear elastic attachments between the cell and the substrate. The evolution of the focal adhesions is coupled to local intracellular stresses which arise from mechanical cell-substrate interactions. Using this model we show that the cell has at least three mechanisms through which it can control its intracellular stresses: focal adhesion position, size, and attachment strength. We also propose that one reason why focal adhesions are typically located on the cell periphery instead of its center is because peripheral focal adhesions allow the cell to be more sensitive to changes in the microenvironment. This increased sensitivity is caused by the fact that peripherally located focal adhesions allow the cells to modulate its intracellular properties over a much larger portion of the cell area.
Collapse
|
205
|
Platet N, Hinkel I, Richert L, Murdamoothoo D, Moufok-Sadoun A, Vanier M, Lavalle P, Gaiddon C, Vautier D, Freund JN, Gross I. The tumor suppressor CDX2 opposes pro-metastatic biomechanical modifications of colon cancer cells through organization of the actin cytoskeleton. Cancer Lett 2017; 386:57-64. [DOI: 10.1016/j.canlet.2016.10.040] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 10/20/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023]
|
206
|
Akalp U, Schnatwinkel C, Stoykovich MP, Bryant SJ, Vernerey FJ. Structural Modeling of Mechanosensitivity in Non-Muscle Cells: Multiscale Approach to Understand Cell Sensing. ACS Biomater Sci Eng 2017; 3:2934-2942. [PMID: 29202009 DOI: 10.1021/acsbiomaterials.6b00693] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The contraction and spreading of nonmuscle cells are important phenomena in a number of cellular processes such as differentiation, morphogenesis, and tissue growth. Recent experimental work has shown that the topology and the mechanical properties of the underlying substrate play a significant role in directing the cell's response. In this work, we introduce a multiscale model to understand the sensing, activation, and contraction of the actin cytoskeleton of nonmuscle cells based on the idea that acto-myosin cross-bridges display a catch-bond response. After investigating the respective roles of bond catchiness and acto-myosin assembly on the mechano-sensitivity of stress fibers, we present full simulations of cells laying on arrays of micropillars. Model predictions show good qualitative agreements with experimental observation, suggesting that acto-myosin catch bonds are a major mechano-sensing element in nonmuscle cells.
Collapse
Affiliation(s)
- Umut Akalp
- Department of Mechanical Engineering, University of Colorado, Boulder, Colorado 80309, United States
| | - Carsten Schnatwinkel
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80309, United States
| | - Mark P Stoykovich
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80309, United States
| | - Stephanie J Bryant
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado 80309, United States
| | - Franck J Vernerey
- Department of Mechanical Engineering, University of Colorado, Boulder, Colorado 80309, United States
| |
Collapse
|
207
|
Tanja Mierke C. Physical role of nuclear and cytoskeletal confinements in cell migration mode selection and switching. AIMS BIOPHYSICS 2017. [DOI: 10.3934/biophy.2017.4.615] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
208
|
Batnyam O, Suye SI, Fujita S. Direct cryopreservation of adherent cells on an elastic nanofiber sheet featuring a low glass-transition temperature. RSC Adv 2017. [DOI: 10.1039/c7ra10604a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Electrospun nanofibers, featured a lower glass-transition temperature than the freezing temperature and a loose mesh structure, allows the direct cryopreservation of adherent cells towards the investigation of cell-material composites.
Collapse
Affiliation(s)
- Onon Batnyam
- Department of Frontier Fiber Technology and Science
- Graduate School of Engineering
- University of Fukui
- Fukui-city
- Japan
| | - Shin-ichiro Suye
- Department of Frontier Fiber Technology and Science
- Graduate School of Engineering
- University of Fukui
- Fukui-city
- Japan
| | - Satoshi Fujita
- Department of Frontier Fiber Technology and Science
- Graduate School of Engineering
- University of Fukui
- Fukui-city
- Japan
| |
Collapse
|
209
|
Park S, Lee S, Park EJ, Kang M, So I, Jeon JH, Chun JN. TGFβ1 induces stress fiber formation through upregulation of TRPC6 in vascular smooth muscle cells. Biochem Biophys Res Commun 2017; 483:129-134. [DOI: 10.1016/j.bbrc.2016.12.179] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 12/27/2016] [Indexed: 01/01/2023]
|
210
|
Livne A, Geiger B. The inner workings of stress fibers - from contractile machinery to focal adhesions and back. J Cell Sci 2016; 129:1293-304. [PMID: 27037413 DOI: 10.1242/jcs.180927] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Ventral stress fibers and focal adhesions are physically coupled structures that play key roles in cellular mechanics and force sensing. The tight functional interdependence between the two is manifested not only by their apparent proximity but also by the fact that ventral stress fibers and focal adhesions are simultaneously diminished upon actomyosin relaxation, and grow when subjected to external stretching. However, whereas the apparent co-regulation of the two structures is well-documented, the underlying mechanisms remains poorly understood. In this Commentary, we discuss some of the fundamental, yet still open questions regarding ventral stress fiber structure, its force-dependent assembly, as well as its capacity to generate force. We also challenge the common approach - i.e. ventral stress fibers are variants of the well-studied striated or smooth muscle machinery - by presenting and critically discussing alternative venues. By highlighting some of the less-explored aspects of the interplay between stress fibers and focal adhesions, we hope that this Commentary will encourage further investigation in this field.
Collapse
Affiliation(s)
- Ariel Livne
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Benjamin Geiger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
211
|
NDN is an imprinted tumor suppressor gene that is downregulated in ovarian cancers through genetic and epigenetic mechanisms. Oncotarget 2016; 7:3018-32. [PMID: 26689988 PMCID: PMC4823087 DOI: 10.18632/oncotarget.6576] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/21/2015] [Indexed: 12/18/2022] Open
Abstract
NDN is a maternally imprinted gene consistently expressed in normal ovarian epithelium, is dramatically downregulated in the majority of ovarian cancers. Little or no NDN expression could be detected in 73% of 351 epithelial ovarian cancers. NDN was also downregulated in 10 ovarian cancer cell lines with total loss in 6 of 10. Re-expression of NDN decreased Bcl-2 levels and induced apoptosis, which significantly inhibited ovarian cancer cell growth in cell culture and in xenografts. In addition, re-expression of NDN inhibited cell migration by decreasing actin stress fiber and focal adhesion complex formation through deactivation of Src, FAK and RhoA. Loss of NDN expression in ovarian cancers could be attributed to LOH in 28% of 18 informative cases and to hypermethylation of CpG sites 1 and 2 of NDN promoter in 23% and 30% of 43 ovarian cancers, respectively. Promoter hypermethylation was also found in 5 of 10 ovarian cancer cell lines. Treatment with the demethylating agent 5-aza-2′-deoxycytidine restored NDN expression in 4 of 7 cell lines with enhanced promoter methylation levels. These observations support the conclusion that NDN is an imprinted tumor suppressor gene which affects cancer cell motility, invasion and growth and that its loss of function in ovarian cancer can be caused by both genetic and epigenetic mechanisms.
Collapse
|
212
|
Annexin A2 is critical for blood-testis barrier integrity and spermatid disengagement in the mammalian testis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:527-545. [PMID: 27974247 DOI: 10.1016/j.bbamcr.2016.12.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 12/06/2016] [Accepted: 12/09/2016] [Indexed: 01/08/2023]
Abstract
Throughout spermatogenesis, two important processes occur at late stage VIII of the seminiferous epithelial cycle in the rat testis: preleptotene spermatocytes commence entry into the adluminal compartment and step 19 spermatids release from the seminiferous epithelium. Presently, it is not clear how these processes, which involve extensive restructuring of unique Sertoli-Sertoli and Sertoli-germ cell junctions, are mediated. We aimed to determine whether annexin A2 (ANXA2), a Ca2+-dependent and phospholipid-binding protein, participates in cell junction dynamics. To address this, in vitro and in vivo RNA interference studies were performed on prepubertal Sertoli cells and adult rat testes. The endpoints of Anxa2 knockdown were determined by immunoblotting, morphological analyses, fluorescent immunostaining, and barrier integrity assays. In the testis, ANXA2 localized to the Sertoli cell stalk, with specific staining at the blood-testis barrier and the concave (ventral) surface of elongated spermatids. ANXA2 also bound actin when testis lysates were used for immunoprecipitation. Anxa2 knockdown was found to disrupt the Sertoli cell/blood-testis barrier in vitro and in vivo. The disruption in barrier function was substantiated by changes in the localization of claudin-11, zona occludens-1, N-cadherin, and β-catenin. Furthermore, Anxa2 knockdown resulted in spermiation defects caused by a dysfunction of tubulobulbar complexes, testis-specific actin-rich ultrastructures that internalize remnant cell junction components prior to spermiation. Additionally, there were changes in the localization of several tubulobulbar complex component proteins, including actin-related protein 3, cortactin, and dynamin I/II. Our results indicate that ANXA2 is critical for the integrity of the blood-testis barrier and the timely release of spermatids.
Collapse
|
213
|
Löffek S, Franzke CW, Helfrich I. Tension in Cancer. Int J Mol Sci 2016; 17:ijms17111910. [PMID: 27854331 PMCID: PMC5133907 DOI: 10.3390/ijms17111910] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/02/2016] [Accepted: 11/09/2016] [Indexed: 12/30/2022] Open
Abstract
Integrins represent a large family of cell receptors that mediate adhesion to the extracellular matrix (ECM), thereby modulating a variety of cellular functions that are required for proliferation, migration, malignant conversion and invasiveness. During tumorigenesis the conversion of a tumor cell from sessile, stationary phenotype to an invasive phenotype requires the ability of tumor cells to interact with their environment in order to transduce signals from the ECM into the cells. Hence, there is increasing evidence that changes in the composition, topography and tension of tumor matrix can be sensed by integrin receptors, leading to the regulation of intracellular signalling events which subsequently help to fuel cancer progression. The fact that intracellular signals perceived from integrin ligand binding impact on almost all steps of tumor progression, including tumor cell proliferation, survival, metastatic dissemination and colonization of a metastatic niche, renders integrins as ideal candidates for the development of therapeutic agents. In this review we summarize the role of integrins in cancer with the special focus on cancer therapies and the recent progress that has been made in the understanding of “integrin-induced tension in cancer”. Finally, we conclude with clinical evidence for the role of integrin-mediated mechanotransduction in the development of therapy-resistant tumors.
Collapse
Affiliation(s)
- Stefanie Löffek
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany.
- German Cancer Consortium (DKTK), University Duisburg-Essen, 45147 Essen, Germany.
| | - Claus-Werner Franzke
- Department of Dermatology and Venerology, Medical Center, University of Freiburg, Hauptstraße 7, 79104 Freiburg, Germany.
| | - Iris Helfrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, 45147 Essen, Germany.
- German Cancer Consortium (DKTK), University Duisburg-Essen, 45147 Essen, Germany.
| |
Collapse
|
214
|
Zhang C, Zhou L, Zhang F, Lü D, Li N, Zheng L, Xu Y, Li Z, Sun S, Long M. Mechanical remodeling of normally sized mammalian cells under a gravity vector. FASEB J 2016; 31:802-813. [PMID: 27871065 DOI: 10.1096/fj.201600897rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 10/31/2016] [Indexed: 01/23/2023]
Abstract
Translocation of the dense nucleus along a gravity vector initiates mechanical remodeling of a cell, but the underlying mechanisms of cytoskeletal network and focal adhesion complex (FAC) reorganization in a mammalian cell remain unclear. We quantified the remodeling of an MC3T3-E1 cell placed in upward-, downward-, or edge-on-orientated substrate. Nucleus longitudinal translocation presents a high value in downward orientation at 24 h or in edge-on orientation at 72 h, which is consistent with orientation-dependent distribution of perinuclear actin stress fibers and vimentin cords. Redistribution of total FAC area and fractionized super mature adhesion number coordinates this dependence at short duration. This orientation-dependent remodeling is associated with nucleus flattering and lamin A/C phosphorylation. Actin depolymerization or Rho-associated protein kinase signaling inhibition abolishes the orientation dependence of nucleus translocation, whereas tubulin polymerization inhibition or vimentin disruption reserves the dependence. A biomechanical model is therefore proposed for integrating the mechanosensing of nucleus translocation with cytoskeletal remodeling and FAC reorganization induced by a gravity vector.-Zhang, C., Zhou, L., Zhang, F., Lü, D., Li, N., Zheng, L., Xu, Y., Li, Z., Sun, S., Long, M. Mechanical remodeling of normally sized mammalian cells under a gravity vector.
Collapse
Affiliation(s)
- Chen Zhang
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Lüwen Zhou
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Fan Zhang
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Dongyuan Lü
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Ning Li
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Lu Zheng
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Yanhong Xu
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Zhan Li
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Shujin Sun
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Mian Long
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
215
|
Exploring the Limits of Cell Adhesion under Shear Stress within Physiological Conditions and beyond on a Chip. Diagnostics (Basel) 2016; 6:diagnostics6040038. [PMID: 27775638 PMCID: PMC5192513 DOI: 10.3390/diagnostics6040038] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/16/2016] [Accepted: 10/13/2016] [Indexed: 12/24/2022] Open
Abstract
Cell adhesion processes are of ubiquitous importance for biomedical applications such as optimization of implant materials. Here, not only physiological conditions such as temperature or pH, but also topographical structures play crucial roles, as inflammatory reactions after surgery can diminish osseointegration. In this study, we systematically investigate cell adhesion under static, dynamic and physiologically relevant conditions employing a lab-on-a-chip system. We screen adhesion of the bone osteosarcoma cell line SaOs-2 on a titanium implant material for pH and temperature values in the physiological range and beyond, to explore the limits of cell adhesion, e.g., for feverish and acidic conditions. A detailed study of different surface roughness Rq gives insight into the correlation between the cells’ abilities to adhere and withstand shear flow and the topography of the substrates, finding a local optimum at Rq = 22 nm. We use shear stress induced by acoustic streaming to determine a measure for the ability of cell adhesion under an external force for various conditions. We find an optimum of cell adhesion for T = 37 °C and pH = 7.4 with decreasing cell adhesion outside the physiological range, especially for high T and low pH. We find constant detachment rates in the physiological regime, but this behavior tends to collapse at the limits of 41 °C and pH 4.
Collapse
|
216
|
Shahriari S, Gordon J, Ghildyal R. Host cytoskeleton in respiratory syncytial virus assembly and budding. Virol J 2016; 13:161. [PMID: 27670781 PMCID: PMC5037899 DOI: 10.1186/s12985-016-0618-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/17/2016] [Indexed: 12/02/2022] Open
Abstract
Respiratory syncytial virus (RSV) is one of the major pathogens responsible for lower respiratory tract infections (LRTI) in young children, the elderly, and the immunosuppressed. Currently, there are no antiviral drugs or vaccines available that effectively target RSV infections, proving a significant challenge in regards to prevention and treatment. An in-depth understanding of the host-virus interactions that underlie assembly and budding would inform new targets for antiviral development.Current research suggests that the polymerised form of actin, the filamentous or F-actin, plays a role in RSV assembly and budding. Treatment with cytochalasin D, which disrupts F-actin, has been shown to inhibit virus release. In addition, the actin cytoskeleton has been shown to interact with the RSV matrix (M) protein, which plays a central role in RSV assembly. For this reason, the interaction between these two components is hypothesised to facilitate the movement of viral components in the cytoplasm and to the budding site. Despite increases in our knowledge of RSV assembly and budding, M-actin interactions are not well understood. In this review, we discuss the current literature on the role of actin cytoskeleton during assembly and budding of RSV with the aim to integrate disparate studies to build a hypothetical model of the various molecular interactions between actin and RSV M protein that facilitate RSV assembly and budding.
Collapse
Affiliation(s)
- Shadi Shahriari
- Respiratory Virology Group, Health Research Institute, Faculty of Education, Science, Technology and Mathematics, University of Canberra, Canberra, 2617, Australia
| | - James Gordon
- Respiratory Virology Group, Health Research Institute, Faculty of Education, Science, Technology and Mathematics, University of Canberra, Canberra, 2617, Australia
| | - Reena Ghildyal
- Respiratory Virology Group, Health Research Institute, Faculty of Education, Science, Technology and Mathematics, University of Canberra, Canberra, 2617, Australia.
| |
Collapse
|
217
|
Abstract
Mechanotransduction is the process through which cells survey the mechanical properties of their environment, convert these mechanical inputs into biochemical signals, and modulate their phenotype in response. These mechanical inputs, which may be encoded in the form of extracellular matrix stiffness, dimensionality, and adhesion, all strongly influence cell morphology, migration, and fate decisions. One mechanism through which cells on planar or pseudo-planar matrices exert tensile forces and interrogate microenvironmental mechanics is through stress fibers, which are bundles composed of actin filaments and, in most cases, non-muscle myosin II filaments. Stress fibers form a continuous structural network that is mechanically coupled to the extracellular matrix through focal adhesions. Furthermore, myosin-driven contractility plays a central role in the ability of stress fibers to sense matrix mechanics and generate tension. Here, we review the distinct roles that non-muscle myosin II plays in driving mechanosensing and focus specifically on motility. In a closely related discussion, we also describe stress fiber classification schemes and the differing roles of various myosin isoforms in each category. Finally, we briefly highlight recent studies exploring mechanosensing in three-dimensional environments, in which matrix content, structure, and mechanics are often tightly interrelated. Stress fibers and the myosin motors therein represent an intriguing and functionally important biological system in which mechanics, biochemistry, and architecture all converge.
Collapse
Affiliation(s)
- Stacey Lee
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, 94720, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, 94720, USA
| |
Collapse
|
218
|
Maninová M, Vomastek T. Dorsal stress fibers, transverse actin arcs, and perinuclear actin fibers form an interconnected network that induces nuclear movement in polarizing fibroblasts. FEBS J 2016; 283:3676-3693. [PMID: 27538255 DOI: 10.1111/febs.13836] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/30/2016] [Accepted: 08/16/2016] [Indexed: 12/12/2022]
Abstract
In polarized motile cells, stress fibers display specific three-dimensional organization. Ventral stress fibers, attached to focal adhesions at both ends, are restricted to the basal side of the cell and nonprotruding cell sides. Dorsal fibers, transverse actin arcs, and perinuclear actin fibers emanate from protruding cell front toward the nucleus and toward apical side of the cell. Perinuclear cap fibers further extend above the nucleus, associate with nuclear envelope through LINC (linker of nucleoskeleton and cytoskeleton) complex and terminate in focal adhesions at cell rear. How are perinuclear actin fibers formed is poorly understood. We show that the formation of perinuclear actin fibers requires dorsal stress fibers that polymerize from focal adhesions at leading edge, and transverse actin arcs that are interconnected with dorsal fibers in spots rich in α-actinin-1. During cell polarization, the interconnected dorsal fibers and transverse arcs move from leading edge toward dorsal side of the cell. As they move, transverse arcs associate with one end of stress fibers present at nonprotruding cell sides, move them above the nucleus thus forming perinuclear actin fibers. Furthermore, the formation of perinuclear actin fibers induces temporal rotational movement of the nucleus resulting in nuclear reorientation to the direction of migration. These results suggest that the network of dorsal fibers, transverse arcs, and perinuclear fibers transfers mechanical signal between the focal adhesions and nuclear envelope that regulates the nuclear reorientation in polarizing cells.
Collapse
Affiliation(s)
| | - Tomáš Vomastek
- Institute of Microbiology AS CR, Prague, Czech Republic.
| |
Collapse
|
219
|
Cheng S, Castillo V, Welty M, Eliaz I, Sliva D. Honokiol inhibits migration of renal cell carcinoma through activation of RhoA/ROCK/MLC signaling pathway. Int J Oncol 2016; 49:1525-1530. [DOI: 10.3892/ijo.2016.3663] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/02/2016] [Indexed: 11/06/2022] Open
|
220
|
Calvo G, Sáenz D, Simian M, Sampayo R, Mamone L, Vallecorsa P, Batlle A, Casas A, Di Venosa G. Reversal of the Migratory and Invasive Phenotype of Ras-Transfected Mammary Cells by Photodynamic Therapy Treatment. J Cell Biochem 2016; 118:464-477. [PMID: 27438675 DOI: 10.1002/jcb.25657] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/19/2016] [Indexed: 12/23/2022]
Abstract
Photodynamic therapy (PDT) is a non-thermal technique for inducing tumor damage following administration of a light-activated photosensitizing drug (PS). In a previous work we found that PDT induces cytoskeleton changes in HB4a-Ras cells (human mammary breast carcinoma HB4a cells transfected with the RAS oncogene). In the present work we have studied the migratory and invasive features and the expression of proteins related to these processes on HB4a-Ras cells after three successive cycles of PDT using different PSs: 5-aminolevulinic acid (ALA), Verteporfin (Verte), m-tetrahydroxyphenylchlorin (m-THPC), and Merocyanine 540 (MC). A slight (1.25- to 2-fold) degree of resistance was acquired in cell populations subjected to the three successive PDT treatments. However, complete cell killing was achieved after a light dose increase. Regardless of the PS employed, all the PDT-treated populations had shorter stress fibres than the untreated control HB4a-Ras cells, and the number of dorsal stress fibres was decreased in the PDT-treated populations. E-Cadherin distribution, which was already aberrant in HB4a-Ras cells, became even more diffuse in the PDT-treated populations, though its expression was increased in some of them. The strong migratory and invasive ability of HB4a-Ras cells in vitro was impaired in all the PDT-treated populations, with a behavior that was similar to the parental non-tumoral HB4a cells. MMP-2 and -9 metalloproteinase activities were also impaired in the PDT-treated populations. The evidence presented herein suggests that the cells surviving PDT would be less metastatic than the initial population. These findings encourage the use of PDT in combination with other treatments such as intraoperative or post-surgery therapeutic procedures. J. Cell. Biochem. 118: 464-477, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Gustavo Calvo
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP), Hospital de Clínicas José de San Martín, CONICET, University of Buenos Aires, Av. Córdoba 2351 1er subsuelo, Ciudad Autónoma de Buenos Aires, Argentina
| | - Daniel Sáenz
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP), Hospital de Clínicas José de San Martín, CONICET, University of Buenos Aires, Av. Córdoba 2351 1er subsuelo, Ciudad Autónoma de Buenos Aires, Argentina
| | - Marina Simian
- Instituto de Nanosistemas & CEDESI, Universidad Nacional de San Martín. 25 de Mayo y Francia, San Martín, Provincia de Buenos Aires, Argentina
| | - Rocío Sampayo
- Instituto de Oncología "Ángel H. Roffo", Av. San Martín 5481, Ciudad Autónoma de Buenos Aires, Argentina
| | - Leandro Mamone
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP), Hospital de Clínicas José de San Martín, CONICET, University of Buenos Aires, Av. Córdoba 2351 1er subsuelo, Ciudad Autónoma de Buenos Aires, Argentina
| | - Pablo Vallecorsa
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP), Hospital de Clínicas José de San Martín, CONICET, University of Buenos Aires, Av. Córdoba 2351 1er subsuelo, Ciudad Autónoma de Buenos Aires, Argentina
| | - Alcira Batlle
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP), Hospital de Clínicas José de San Martín, CONICET, University of Buenos Aires, Av. Córdoba 2351 1er subsuelo, Ciudad Autónoma de Buenos Aires, Argentina
| | - Adriana Casas
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP), Hospital de Clínicas José de San Martín, CONICET, University of Buenos Aires, Av. Córdoba 2351 1er subsuelo, Ciudad Autónoma de Buenos Aires, Argentina
| | - Gabriela Di Venosa
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP), Hospital de Clínicas José de San Martín, CONICET, University of Buenos Aires, Av. Córdoba 2351 1er subsuelo, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
221
|
Khan S, Conte I, Carter T, Bayer KU, Molloy JE. Multiple CaMKII Binding Modes to the Actin Cytoskeleton Revealed by Single-Molecule Imaging. Biophys J 2016; 111:395-408. [PMID: 27463141 PMCID: PMC4968397 DOI: 10.1016/j.bpj.2016.06.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 11/22/2022] Open
Abstract
Localization of the Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) to dendritic spine synapses is determined in part by the actin cytoskeleton. We determined binding of GFP-tagged CaMKII to tag-RFP-labeled actin cytoskeleton within live cells using total internal reflection fluorescence microscopy and single-molecule tracking. Stepwise photobleaching showed that CaMKII formed oligomeric complexes. Photoactivation experiments demonstrated that diffusion out of the evanescent field determined the track lifetimes. Latrunculin treatment triggered a coupled loss of actin stress fibers and the colocalized, long-lived CaMKII tracks. The CaMKIIα (α) isoform, which was previously thought to lack F-actin interactions, also showed binding, but this was threefold weaker than that observed for CaMKIIβ (β). The βE' splice variant bound more weakly than α, showing that binding by β depends critically on the interdomain linker. The mutations βT287D and αT286D, which mimic autophosphorylation states, also abolished F-actin binding. Autophosphorylation triggers autonomous CaMKII activity, but does not impair GluN2B binding, another important synaptic protein interaction of CaMKII. The CaMKII inhibitor tatCN21 or CaMKII mutations that inhibit GluN2B association by blocking binding of ATP (βK43R and αK42M) or Ca(2+)/calmodulin (βA303R) had no effect on the interaction with F-actin. These results provide the first rationale for the reduced synaptic spine localization of the αT286D mutant, indicating that transient F-actin binding contributes to the synaptic localization of the CaMKIIα isoform. The track lifetime distributions had a stretched exponential form consistent with a heterogeneously diffusing population. This heterogeneity suggests that CaMKII adopts different F-actin binding modes, which is most easily rationalized by multiple subunit contacts between the CaMKII dodecamer and the F-actin cytoskeleton that stabilize the initial weak (micromolar) monovalent interaction.
Collapse
Affiliation(s)
- Shahid Khan
- Molecular Biology Consortium, Lawrence Berkeley National Laboratory, Berkeley, California.
| | - Ianina Conte
- Cardiovascular and Cell Science Research Institute, St. George's University of London, London, UK
| | - Tom Carter
- Cell Biology and Genetics, St. George's University of London, London, UK
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado
| | - Justin E Molloy
- The Francis Crick Institute, Mill Hill Laboratory, London, UK
| |
Collapse
|
222
|
Kundu M, Mahata B, Banerjee A, Chakraborty S, Debnath S, Ray SS, Ghosh Z, Biswas K. Ganglioside GM2 mediates migration of tumor cells by interacting with integrin and modulating the downstream signaling pathway. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1863:1472-1489. [PMID: 27066976 DOI: 10.1016/j.bbamcr.2016.04.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 04/01/2016] [Accepted: 04/06/2016] [Indexed: 11/19/2022]
Abstract
The definitive role of ganglioside GM2 in mediating tumor-induced growth and progression is still unknown. Here we report a novel role of ganglioside GM2 in mediating tumor cell migration and uncovered its mechanism. Data shows differential expression levels of GM2-synthase as well as GM2 in different human cancer cells. siRNA mediated knockdown of GM2-synthase in CCF52, A549 and SK-RC-26B cells resulted in significant inhibition of tumor cell migration as well as invasion in vitro without affecting cellular proliferation. Over-expression of GM2-synthase in low-GM2 expressing SK-RC-45 cells resulted in a consequent increase in migration thus confirming the potential role GM2 and its downstream partners play in tumor cell migration and motility. Further, treatment of SK-RC-45 cells with exogenous GM2 resulted in a dramatic increase in migratory and invasive capacity with no change in proliferative capacity, thereby confirming the role of GM2 in tumorigenesis specifically by mediating tumor migration and invasion. Gene expression profiling of GM2-synthase silenced cells revealed altered expression of several genes involved in cell migration primarily those controlling the integrin mediated signaling. GM2-synthase knockdown resulted in decreased phosphorylation of FAK, Src as well as Erk, while over-expression and/or exogenous GM2 treatment caused increased FAK and Erk phosphorylation respectively. Again, GM2 mediated invasion and Erk phosphorylation is blocked in integrin knockdown SK-RC-45 cells, thus confirming that GM2 mediated migration and phosphorylation of Erk is integrin dependent. Finally, confocal microscopy suggested co-localization while co-immunoprecipitation and surface plasmon resonance (SPR) confirmed direct interaction of membrane bound ganglioside, GM2 with the integrin receptor.
Collapse
Affiliation(s)
- Manjari Kundu
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal 700054, India
| | - Barun Mahata
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal 700054, India
| | - Avisek Banerjee
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal 700054, India
| | - Sohini Chakraborty
- The Bioinformatics Center, Bose Institute, Kolkata, West Bengal 700054, India
| | - Shibjyoti Debnath
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal 700054, India
| | | | - Zhumur Ghosh
- The Bioinformatics Center, Bose Institute, Kolkata, West Bengal 700054, India
| | - Kaushik Biswas
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal 700054, India.
| |
Collapse
|
223
|
Vernerey FJ, Akalp U. Role of catch bonds in actomyosin mechanics and cell mechanosensitivity. Phys Rev E 2016; 94:012403. [PMID: 27575160 PMCID: PMC5542069 DOI: 10.1103/physreve.94.012403] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Indexed: 01/10/2023]
Abstract
We propose a mechanism of adherent cell mechanosensing, based on the idea that the contractile actomyosin machinery behaves as a catch bond. For this, we construct a simplified model of the actomyosin structure that constitutes the building block of stress fibers and express the stability of cross bridges in terms of the force-dependent bonding energy of the actomyosin bond. Consistent with experimental measurements, we then consider that the energy barrier of the actomyosin bond increases for tension and show that this response is enough to explain the force-induced stabilization of a stress fiber. Further numerical simulations at the cellular level show that the catch-bond hypothesis can help in understanding and predict the sensitivity of adherent cells to substrate stiffness.
Collapse
Affiliation(s)
- Franck J. Vernerey
- Department of Mechanical Engineering, University of Colorado, Boulder, Colorado 80309, USA
| | - Umut Akalp
- Department of Mechanical Engineering, University of Colorado, Boulder, Colorado 80309, USA
| |
Collapse
|
224
|
García Ponce A, Citalán Madrid AF, Vargas Robles H, Chánez Paredes S, Nava P, Betanzos A, Zarbock A, Rottner K, Vestweber D, Schnoor M. Loss of cortactin causes endothelial barrier dysfunction via disturbed adrenomedullin secretion and actomyosin contractility. Sci Rep 2016; 6:29003. [PMID: 27357373 PMCID: PMC4928053 DOI: 10.1038/srep29003] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 06/13/2016] [Indexed: 12/28/2022] Open
Abstract
Changes in vascular permeability occur during inflammation and the actin cytoskeleton plays a crucial role in regulating endothelial cell contacts and permeability. We demonstrated recently that the actin-binding protein cortactin regulates vascular permeability via Rap1. However, it is unknown if the actin cytoskeleton contributes to increased vascular permeability without cortactin. As we consistently observed more actin fibres in cortactin-depleted endothelial cells, we hypothesised that cortactin depletion results in increased stress fibre contractility and endothelial barrier destabilisation. Analysing the contractile machinery, we found increased ROCK1 protein levels in cortactin-depleted endothelium. Concomitantly, myosin light chain phosphorylation was increased while cofilin, mDia and ERM were unaffected. Secretion of the barrier-stabilising hormone adrenomedullin, which activates Rap1 and counteracts actomyosin contractility, was reduced in plasma from cortactin-deficient mice and in supernatants of cortactin-depleted endothelium. Importantly, adrenomedullin administration and ROCK1 inhibition reduced actomyosin contractility and rescued the effect on permeability provoked by cortactin deficiency in vitro and in vivo. Our data suggest a new role for cortactin in controlling actomyosin contractility with consequences for endothelial barrier integrity.
Collapse
Affiliation(s)
- Alexander García Ponce
- Department for Molecular Biomedicine, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| | - Alí F Citalán Madrid
- Department for Molecular Biomedicine, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| | - Hilda Vargas Robles
- Department for Molecular Biomedicine, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| | - Sandra Chánez Paredes
- Department for Molecular Biomedicine, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| | - Porfirio Nava
- Department for Physiology, Biophysics and Neurosciences, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| | - Abigail Betanzos
- Department for Infectomics and Molecular Pathogenesis, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Clinic of Münster, 48149 Münster, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, TU Braunschweig, 38106 Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Dietmar Vestweber
- Department for Vascular Cell Biology, Max-Planck-Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Michael Schnoor
- Department for Molecular Biomedicine, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| |
Collapse
|
225
|
The actin crosslinking protein palladin modulates force generation and mechanosensitivity of tumor associated fibroblasts. Sci Rep 2016; 6:28805. [PMID: 27353427 PMCID: PMC4926206 DOI: 10.1038/srep28805] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 06/10/2016] [Indexed: 01/15/2023] Open
Abstract
Cells organize actin filaments into higher-order structures by regulating the composition, distribution and concentration of actin crosslinkers. Palladin is an actin crosslinker found in the lamellar actin network and stress fibers, which are critical for mechanosensing of the environment. Palladin also serves as a molecular scaffold for α-actinin, another key actin crosslinker. By virtue of its close interactions with actomyosin structures in the cell, palladin may play an important role in cell mechanics. However, the role of palladin in cellular force generation and mechanosensing has not been studied. Here, we investigate the role of palladin in regulating the plasticity of the actin cytoskeleton and cellular force generation in response to alterations in substrate stiffness. Traction force microscopy revealed that tumor-associated fibroblasts generate larger forces on substrates of increased stiffness. Contrary to expectations, knocking down palladin increased the forces generated by cells and inhibited their ability to sense substrate stiffness for very stiff gels. This was accompanied by significant differences in actin organization, adhesion dynamics and altered myosin organization in palladin knock-down cells. Our results suggest that actin crosslinkers such as palladin and myosin motors coordinate for optimal cell function and to prevent aberrant behavior as in cancer metastasis.
Collapse
|
226
|
Shang Y, Wang H, Jia P, Zhao H, Liu C, Liu W, Song Z, Xu Z, Yang L, Wang Y, Li W. Autophagy regulates spermatid differentiation via degradation of PDLIM1. Autophagy 2016; 12:1575-92. [PMID: 27310465 DOI: 10.1080/15548627.2016.1192750] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Spermiogenesis is a complex and highly ordered spermatid differentiation process that requires reorganization of cellular structures. We have previously found that Atg7 is required for acrosome biogenesis. Here, we show that autophagy regulates the round and elongating spermatids. Specifically, we found that Atg7 is required for spermatozoa flagella biogenesis and cytoplasm removal during spermiogenesis. Spermatozoa motility of atg7-null mice dropped significantly with some extra-cytoplasm retained on the mature sperm head. These defects are associated with an impairment of the cytoskeleton organization. Functional screening revealed that the negative cytoskeleton organization regulator, PDLIM1 (PDZ and LIM domain 1 [elfin]), needs to be degraded by the autophagy-lysosome-dependent pathway to facilitate the proper organization of the cytoskeleton. Our results thus provide a novel mechanism showing that autophagy regulates cytoskeleton organization mainly via degradation of PDLIM1 to facilitate the differentiation of spermatids.
Collapse
Affiliation(s)
- Yongliang Shang
- a State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China.,b University of Chinese Academy of Sciences , Beijing , China
| | - Hongna Wang
- a State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China.,b University of Chinese Academy of Sciences , Beijing , China
| | - Pengfei Jia
- c State Key Laboratory of Molecular Developmental Biology and National Center for Plant Gene Research (Beijing), Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing , China
| | - Haichao Zhao
- a State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China.,b University of Chinese Academy of Sciences , Beijing , China
| | - Chao Liu
- a State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China.,b University of Chinese Academy of Sciences , Beijing , China
| | - Weixiao Liu
- a State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Zhenhua Song
- a State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China.,b University of Chinese Academy of Sciences , Beijing , China
| | - Zhiliang Xu
- a State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China.,b University of Chinese Academy of Sciences , Beijing , China
| | - Lin Yang
- c State Key Laboratory of Molecular Developmental Biology and National Center for Plant Gene Research (Beijing), Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing , China
| | - Yanfang Wang
- d State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences , Beijing , China
| | - Wei Li
- a State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| |
Collapse
|
227
|
Lv F, Zhu L, Zhang J, Yu J, Cheng X, Peng B. Evaluation of the in vitro
biocompatibility of a new fast-setting ready-to-use root filling and repair material. Int Endod J 2016; 50:540-548. [PMID: 27214303 DOI: 10.1111/iej.12661] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 05/17/2016] [Indexed: 11/29/2022]
Affiliation(s)
- F. Lv
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - L. Zhu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - J. Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - J. Yu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - X. Cheng
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
| | - B. Peng
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan China
| |
Collapse
|
228
|
Hensley MR, Cui Z, Chua RFM, Simpson S, Shammas NL, Yang JY, Leung YF, Zhang G. Evolutionary and developmental analysis reveals KANK genes were co-opted for vertebrate vascular development. Sci Rep 2016; 6:27816. [PMID: 27292017 PMCID: PMC4904190 DOI: 10.1038/srep27816] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/25/2016] [Indexed: 11/28/2022] Open
Abstract
Gene co-option, usually after gene duplication, in the evolution of development is found to contribute to vertebrate morphological innovations, including the endothelium-based vascular system. Recently, a zebrafish kank gene was found expressed in the vascular vessel primordium, suggesting KANK genes are a component of the developmental tool kit for the vertebrate vascular system. However, how the KANK gene family is involved in vascular vessel development during evolution remains largely unknown. First, we analyzed the molecular evolution of the KANK genes in metazoan, and found that KANK1, KANK2, KANK3 and KANK4 emerged in the lineage of vertebrate, consistent with the two rounds of vertebrate whole-genome duplications (WGD). Moreover, KANK genes were further duplicated in teleosts through the bony-fish specific WGD, while only kank1 and kank4 duplicates were retained in some of the examined fish species. We also found all zebrafish kank genes, except kank1b, are primarily expressed during embryonic vascular development. Compared to invertebrate KANK gene expression in the central nervous system, the vascular expression of zebrafish kank genes suggested KANK genes were co-opted for vertebrate vascular development. Given the cellular roles of KANK genes, our results suggest that this co-option may facilitate the evolutionary origin of vertebrate vascular vessels.
Collapse
Affiliation(s)
- Monica R Hensley
- Department of Comparative Pathobiology;.725 Harrison Street, West Lafayette, IN 47907, USA
| | - Zhibin Cui
- Department of Comparative Pathobiology;.725 Harrison Street, West Lafayette, IN 47907, USA
| | - Rhys F M Chua
- Department of Comparative Pathobiology;.725 Harrison Street, West Lafayette, IN 47907, USA
| | - Stefanie Simpson
- Department of Comparative Pathobiology;.725 Harrison Street, West Lafayette, IN 47907, USA
| | - Nicole L Shammas
- Department of Comparative Pathobiology;.725 Harrison Street, West Lafayette, IN 47907, USA
| | - Jer-Yen Yang
- Purdue University Center for Cancer Research; 725 Harrison Street, West Lafayette, IN 47907, USA.,Department of Basic Medical Sciences; 725 Harrison Street, West Lafayette, IN 47907,USA
| | - Yuk Fai Leung
- Department of Biological Sciences, 725 Harrison Street, West Lafayette, IN 47907,USA.,Integrative Neuroscience Center; 725 Harrison Street, West Lafayette, IN 47907, USA
| | - GuangJun Zhang
- Department of Comparative Pathobiology;.725 Harrison Street, West Lafayette, IN 47907, USA.,Purdue University Center for Cancer Research; 725 Harrison Street, West Lafayette, IN 47907, USA.,Integrative Neuroscience Center; 725 Harrison Street, West Lafayette, IN 47907, USA.,Purdue Institute for Inflammation, Immunology and Infectious Diseases (PI4D), 725 Harrison Street, West Lafayette, IN 47907, USA
| |
Collapse
|
229
|
Hiebl B, Cui J, Kratz K, Frank O, Schossig M, Richau K, Lee S, Jung F, Lendlein A. Viability, morphology and function of primary endothelial cells on poly(n-butyl acrylate) networks having elastic moduli comparable to arteries. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2016; 23:901-15. [PMID: 21457619 DOI: 10.1163/092050611x566144] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Soft hydrophobic poly(n-butyl acrylate) networks (cPnBA) were developed as entropy elastic substrates for passive mechanical stimulation of cells, where the elastic modulus of the cPnBAs could be systematically adjusted by variation of the cross-link density. The networks were synthesized by thermally-induced radical polymerization from n-butyl acrylate, with poly(propylene glycol) dimethacrylate (PPGDMA) acting as cross-linker, whereby the purity of the cPnBAs was confirmed by(1) H-NMR spectroscopy and gas chromatography. In this work two cPnBA polymer networks with an elastic modulus around 200 kPa and 1 MPa were investigated having an elastic modulus similar to that of arteries. Both cPnBAs exhibited an almost smooth surface with a surface roughness (R q) in the wet state ranging from 17 to 37 nm and a similar zetapotential, indicating an almost identical chemical composition within the topmost surface layer in terms of functional groups. In contrast, wettability of the samples was found to be different with an advancing angle ( advancing) of 123 ± 3.8° for cPnBA0250, while for cPnBA1100 significantly lower values for advancing (111 ± 3.8°) were obtained. First in vitro tests were performed with primary endothelial cells (HUVEC) to study its effects on vascular cell functions. Within the time period of cultivation (72 h), the cells on the cPnBA samples reached subconfluence and showed a viability rate of almost 100%. Although cell density differed after 72 h with more cells on cPnBA0250 than on cPnBA1100, both materials showed no significant effect on the cell morphology, the cellular LDH-release, which was used as marker for the integrity of the cell membrane, and the organisation of the VE-cadherin. However, lower cell density and less actin stress fibre formation on cPnBA1100 might indicate that cell-material interaction was weaker on cPnBA1100 than on cPnBA0250. The secretion of the vasoactive cytokines prostacyclin (PGI2) and thromboxane A2 (TXA2) was low compared to previously reported values. However, the anti-thrombogenic ratio of PGI2/TXA2 - which is balanced under physiological conditions - with much higher PGI2 compared to TXA2 (up to 17.6-fold after 72 h for cPnBA1100) suggests that this material might be effective to preventing thrombosis.
Collapse
Affiliation(s)
- B Hiebl
- a Center for Biomaterial Development, Institute of Polymer Research, Helmholtz-Zentrum Geesthacht, Teltow, Germany; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Nagahama M, Ohkubo A, Kinouchi Y, Kobayashi K, Miyamoto K, Takehara M, Sakurai J. Clostridium perfringens TpeL Induces Formation of Stress Fibers via Activation of RhoA-ROCK Signaling Pathway. Biol Pharm Bull 2016; 38:732-9. [PMID: 25947919 DOI: 10.1248/bpb.b14-00842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Clostridium perfringens TpeL belongs to a family of large clostridial glucosylating cytotoxins. TpeL modifies Rac1 and Ras subfamily proteins. Herein we report TpeL-induced formation of stress fibers via RhoA-Rho kinase (ROCK) signaling. A recombinant protein (TpeL1-525) derived from the TpeL N-terminal catalytic domain in the presence of streptolysin O (SLO) induced the formation of actin stress fibers in Madin-Darby canine kidney (MDCK) cells in a dose-dependent manner. The RhoA/ROCK pathway is known to control the formation of stress fibers. We examined the role of the RhoA/ROCK pathway in TpeL-induced formation of stress fibers. TpeL1-525-induced formation of stress fibers was inhibited by the ROCK inhibitor, Y27632 and Rho protein inhibitor, C3 transferase. TpeL1-525 activated RhoA and ROCK in a dose-dependent manner. C3 transferase blocked TpeL1-525-induced activation of RhoA and ROCK whereas Y27632 inhibited TpeL-induced activation of ROCK. These results demonstrate for the first time that TpeL induces the formation of stress fibers by activating the RhoA/ROCK signaling pathway.
Collapse
Affiliation(s)
- Masahiro Nagahama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University
| | | | | | | | | | | | | |
Collapse
|
231
|
Kronlage C, Schäfer-Herte M, Böning D, Oberleithner H, Fels J. Feeling for Filaments: Quantification of the Cortical Actin Web in Live Vascular Endothelium. Biophys J 2016; 109:687-98. [PMID: 26287621 PMCID: PMC4547164 DOI: 10.1016/j.bpj.2015.06.066] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 06/07/2015] [Accepted: 06/24/2015] [Indexed: 12/27/2022] Open
Abstract
Contact-mode atomic force microscopy (AFM) has been shown to reveal cortical actin structures. Using live endothelial cells, we visualized cortical actin dynamics simultaneously by AFM and confocal fluorescence microscopy. We present a method that quantifies dynamic changes in the mechanical ultrastructure of the cortical actin web. We argue that the commonly used, so-called error signal imaging in AFM allows a qualitative, but not quantitative, analysis of cortical actin dynamics. The approach we used comprises fast force-curve-based topography imaging and subsequent image processing that enhances local height differences. Dynamic changes in the organization of the cytoskeleton network can be observed and quantified by surface roughness calculations and automated morphometrics. Upon treatment with low concentrations of the actin-destabilizing agent cytochalasin D, the cortical cytoskeleton network is thinned out and the average mesh size increases. In contrast, jasplakinolide, a drug that enhances actin polymerization, consolidates the cytoskeleton network and reduces the average mesh area. In conclusion, cortical actin dynamics can be quantified in live cells. To our knowledge, this opens a new pathway for conducting quantitative structure-function analyses of the endothelial actin web just beneath the apical plasma membrane.
Collapse
Affiliation(s)
| | - Marco Schäfer-Herte
- Institute of Cell Dynamics and Imaging, University of Münster, Münster, Germany
| | - Daniel Böning
- Institute of Medical Physics and Biophysics, University of Münster, Münster, Germany
| | | | - Johannes Fels
- Institute of Physiology II, University of Münster, Münster, Germany; Institute of Cell Dynamics and Imaging, University of Münster, Münster, Germany.
| |
Collapse
|
232
|
Gandalovičová A, Vomastek T, Rosel D, Brábek J. Cell polarity signaling in the plasticity of cancer cell invasiveness. Oncotarget 2016; 7:25022-49. [PMID: 26872368 PMCID: PMC5041887 DOI: 10.18632/oncotarget.7214] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 01/29/2016] [Indexed: 02/07/2023] Open
Abstract
Apico-basal polarity is typical of cells present in differentiated epithelium while front-rear polarity develops in motile cells. In cancer development, the transition from epithelial to migratory polarity may be seen as the hallmark of cancer progression to an invasive and metastatic disease. Despite the morphological and functional dissimilarity, both epithelial and migratory polarity are controlled by a common set of polarity complexes Par, Scribble and Crumbs, phosphoinositides, and small Rho GTPases Rac, Rho and Cdc42. In epithelial tissues, their mutual interplay ensures apico-basal and planar cell polarity. Accordingly, altered functions of these polarity determinants lead to disrupted cell-cell adhesions, cytoskeleton rearrangements and overall loss of epithelial homeostasis. Polarity proteins are further engaged in diverse interactions that promote the establishment of front-rear polarity, and they help cancer cells to adopt different invasion modes. Invading cancer cells can employ either the collective, mesenchymal or amoeboid invasion modes or actively switch between them and gain intermediate phenotypes. Elucidation of the role of polarity proteins during these invasion modes and the associated transitions is a necessary step towards understanding the complex problem of metastasis. In this review we summarize the current knowledge of the role of cell polarity signaling in the plasticity of cancer cell invasiveness.
Collapse
Affiliation(s)
- Aneta Gandalovičová
- Department of Cell Biology, Charles University in Prague, Viničná, Prague, Czech Republic
| | - Tomáš Vomastek
- Institute of Microbiology, Academy of Sciences of The Czech Republic, Videňská, Prague, Czech Republic
| | - Daniel Rosel
- Department of Cell Biology, Charles University in Prague, Viničná, Prague, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University in Prague, Viničná, Prague, Czech Republic
| |
Collapse
|
233
|
Popov K, Komianos J, Papoian GA. MEDYAN: Mechanochemical Simulations of Contraction and Polarity Alignment in Actomyosin Networks. PLoS Comput Biol 2016; 12:e1004877. [PMID: 27120189 PMCID: PMC4847874 DOI: 10.1371/journal.pcbi.1004877] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 03/22/2016] [Indexed: 11/29/2022] Open
Abstract
Active matter systems, and in particular the cell cytoskeleton, exhibit complex mechanochemical dynamics that are still not well understood. While prior computational models of cytoskeletal dynamics have lead to many conceptual insights, an important niche still needs to be filled with a high-resolution structural modeling framework, which includes a minimally-complete set of cytoskeletal chemistries, stochastically treats reaction and diffusion processes in three spatial dimensions, accurately and efficiently describes mechanical deformations of the filamentous network under stresses generated by molecular motors, and deeply couples mechanics and chemistry at high spatial resolution. To address this need, we propose a novel reactive coarse-grained force field, as well as a publicly available software package, named the Mechanochemical Dynamics of Active Networks (MEDYAN), for simulating active network evolution and dynamics (available at www.medyan.org). This model can be used to study the non-linear, far from equilibrium processes in active matter systems, in particular, comprised of interacting semi-flexible polymers embedded in a solution with complex reaction-diffusion processes. In this work, we applied MEDYAN to investigate a contractile actomyosin network consisting of actin filaments, alpha-actinin cross-linking proteins, and non-muscle myosin IIA mini-filaments. We found that these systems undergo a switch-like transition in simulations from a random network to ordered, bundled structures when cross-linker concentration is increased above a threshold value, inducing contraction driven by myosin II mini-filaments. Our simulations also show how myosin II mini-filaments, in tandem with cross-linkers, can produce a range of actin filament polarity distributions and alignment, which is crucially dependent on the rate of actin filament turnover and the actin filament’s resulting super-diffusive behavior in the actomyosin-cross-linker system. We discuss the biological implications of these findings for the arc formation in lamellipodium-to-lamellum architectural remodeling. Lastly, our simulations produce force-dependent accumulation of myosin II, which is thought to be responsible for their mechanosensation ability, also spontaneously generating myosin II concentration gradients in the solution phase of the simulation volume. Active matter systems have the distinct ability to convert energy from their surroundings into mechanical work, which gives rise to them having highly dynamic properties. Modeling active matter systems and capturing their complex behavior has been a great challenge in past years due to the many coupled interactions between their constituent parts, including not only distinct chemical and mechanical properties, but also feedback between them. One of the most intriguing biological active matter systems is the cell cytoskeleton, which can dynamically respond to chemical and mechanical cues to control cell structure and shape, playing a central role in many higher-order cellular processes. To model these systems and reproduce their behavior, we present a new modeling approach which combines the chemical, mechanical, and molecular transport aspects of active matter systems, all represented with equivalent complexity, while also allowing for various forms of mechanochemical feedback. This modeling approach, named MEDYAN, and software implementation is flexible so that a wide range of active matter systems can be simulated with a high level of detail, and ultimately can help to describe active matter phenomena, and in particular, the dynamics of the cell cytoskeleton. In this work, we have used MEDYAN to simulate a cytoskeletal network consisting of actin filaments, cross-linking proteins, and myosin II molecular motors. We found that these systems show rich dynamical behaviors, undergoing alignment and bundling transitions, with an emergent contractility, as the concentrations of myosin II and cross-linking proteins, as well as actin filament turnover rates, are varied.
Collapse
Affiliation(s)
- Konstantin Popov
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, United States of America
- Institute for Physical Science and Technology, University of Maryland, College Park, Maryland, United States of America
| | - James Komianos
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, United States of America
- Institute for Physical Science and Technology, University of Maryland, College Park, Maryland, United States of America
- Biophysics Graduate Program, University of Maryland, College Park, Maryland, United States of America
| | - Garegin A. Papoian
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, United States of America
- Institute for Physical Science and Technology, University of Maryland, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
234
|
Goryachev AB, Leda M, Miller AL, von Dassow G, Bement WM. How to make a static cytokinetic furrow out of traveling excitable waves. Small GTPases 2016; 7:65-70. [PMID: 27070950 PMCID: PMC4905281 DOI: 10.1080/21541248.2016.1168505] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Emergence of the cytokinetic Rho zone that orchestrates formation and ingression of the cleavage furrow had been explained previously via microtubule-dependent cortical concentration of Ect2, a guanine nucleotide exchange factor for Rho. The results of a recent publication now demonstrate that, en route from resting cortex to fully established furrow, there lies a regime of cortical excitability in which Rho activity and F-actin play the roles of the prototypical activator and inhibitor, respectively. This cortical excitability is manifest as dramatic traveling waves on the cortex of oocytes and embryos of frogs and starfish. These waves are initiated by autocatalytic activation of Rho at the wave front and extinguished by F-actin-dependent inhibition at their back. It is still unclear how propagating excitable Rho-actin waves give rise to the stable co-existence of Rho activity and F-actin density in the static cleavage furrow during cytokinesis. It is possible that some central spindle-associated signaling molecule simply turns off the inhibition of Rho activity by F-actin. However, mathematical modeling suggests a distinct scenario in which local “re-wiring” of the Rho-actin coupling in the furrow is no longer necessary. Instead, the model predicts that the continuously rising level of Ect2 produces in the furrow a qualitatively new stable steady state that replaces excitability and brings about the stable co-existence of high Rho activity and dense F-actin despite the continuing inhibition of Rho by F-actin.
Collapse
Affiliation(s)
- Andrew B Goryachev
- a Centre for Systems and Synthetic Biology, Cell Biology Institute, School of Biological Sciences, University of Edinburgh , Edinburgh , UK
| | - Marcin Leda
- a Centre for Systems and Synthetic Biology, Cell Biology Institute, School of Biological Sciences, University of Edinburgh , Edinburgh , UK
| | - Ann L Miller
- b Department of Molecular , Cellular and Developmental Biology, University of Michigan , Ann-Arbor , MI , USA
| | - George von Dassow
- c Oregon Institute of Marine Biology, University of Oregon , Charleston , OR , USA
| | - William M Bement
- d Laboratory of Cell and Molecular Biology, Graduate Program in Cell and Molecular Biology, University of Wisconsin-Madison , Madison , WI , USA
| |
Collapse
|
235
|
Zhang X, Hu X, Lei H, Hu J, Zhang Y. Mechanical force-induced polymerization and depolymerization of F-actin at water/solid interfaces. NANOSCALE 2016; 8:6008-6013. [PMID: 26928199 DOI: 10.1039/c5nr08713a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Actin molecules are among the three main cytoskeleton proteins of cells and undergo rapid cycling to regulate critical processes such as endocytosis, cytokinesis, cell polarity, and cell morphogenesis. Although extensive studies have been carried out on the dynamics as well as biological functions of actin polymerization and depolymerization both in vivo and in vitro, the molecular mechanisms by which cells sense and respond to mechanical signals are not fully understood. In particular, little attention has been paid to the effect of a physical force that is exerted directly on the actin cytoskeleton. In this paper, we have explored how the mechanical force affects the actin polymerization and depolymerization behaviors at water/solid interfaces using an atomic force microscope (AFM) operated in liquid. By raster scanning an AFM probe on a substrate surface with a certain load, it was found that actin monomers could polymerize into filaments without the help of actin related proteins (ARPs). Further study indicated that actin monomers were inclined to form filaments only under a small scanning load. The polymerized actin filaments would be depolymerized when the mechanical force was stronger. A possible mechanism has been suggested to explain the mechanical force induced actin polymerization.
Collapse
Affiliation(s)
- Xueqiang Zhang
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.
| | | | | | | | | |
Collapse
|
236
|
Kushida N, Yamaguchi O, Kawashima Y, Akaihata H, Hata J, Ishibashi K, Aikawa K, Kojima Y. Uni-axial stretch induces actin stress fiber reorganization and activates c-Jun NH2 terminal kinase via RhoA and Rho kinase in human bladder smooth muscle cells. BMC Urol 2016; 16:9. [PMID: 26928204 PMCID: PMC4772493 DOI: 10.1186/s12894-016-0127-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 02/23/2016] [Indexed: 01/10/2023] Open
Abstract
Background Excessive mechanical overload may be involved in bladder wall remodelling. Since the activity of Rho kinase is known to be upregulated in the obstructed bladder, we investigate the roles of the RhoA/Rho kinase pathway in mechanical overloaded bladder smooth muscle cells. Methods Human bladder smooth muscle cells were stimulated on silicon culture plates by 15 % elongated uni-axial cyclic stretch at 1 Hz. The activity of c-Jun NH2-terminal kinase was measured by western blotting and actin stress fibers were observed by stained with phallotoxin conjugated with Alexa-Fluor 594. Results The activity of c-Jun NH2-terminal kinase 1 peaked at 30 min (4.7-fold increase vs. before stretch) and this activity was partially abrogated by the RhoA inhibitor, C3 exoenzoyme or by the Rho kinase inhibitor, Y-27632. Stretch induced the strong formation of actin stress fibers and these fibers re-orientated in a direction that was perpendicular to the stretch direction. The average angle of the fibers from the perpendicular to the direction of stretch was significantly different between before, and 4 h after, stretch. Actin stress fibers reorganization was also suppressed by the C3 exoenzyme or Y-27632. Conclusions Bladder smooth muscle cells appear to have elaborate mechanisms for sensing mechanical stress and for adapting to mechanical stress overload by cytoskeletal remodeling and by activating cell growth signals such as c-Jun NH2-terminal kinase via RhoA/Rho kinase pathways.
Collapse
Affiliation(s)
- Nobuhiro Kushida
- Department of Urology, Fukushima Medical University School of Medicine, 1, Hikarigaoka, Fukushima, 960-1295, Japan.
| | - Osamu Yamaguchi
- Division of Bioengineering and LUTD Research, Nihon University School of Engineering, Nihon University, 1, Nakagawara, Tokusada, Tamura, Koriyama, 963-8642, Japan.
| | - Yohei Kawashima
- Department of Urology, Fukushima Medical University School of Medicine, 1, Hikarigaoka, Fukushima, 960-1295, Japan.
| | - Hidenori Akaihata
- Department of Urology, Fukushima Medical University School of Medicine, 1, Hikarigaoka, Fukushima, 960-1295, Japan.
| | - Junya Hata
- Department of Urology, Fukushima Medical University School of Medicine, 1, Hikarigaoka, Fukushima, 960-1295, Japan.
| | - Kei Ishibashi
- Department of Urology, Fukushima Medical University School of Medicine, 1, Hikarigaoka, Fukushima, 960-1295, Japan.
| | - Ken Aikawa
- Department of Urology, Fukushima Medical University School of Medicine, 1, Hikarigaoka, Fukushima, 960-1295, Japan.
| | - Yoshiyuki Kojima
- Department of Urology, Fukushima Medical University School of Medicine, 1, Hikarigaoka, Fukushima, 960-1295, Japan.
| |
Collapse
|
237
|
Fallqvist B. Implementing cell contractility in filament-based cytoskeletal models. Cytoskeleton (Hoboken) 2016; 73:93-106. [PMID: 26899417 DOI: 10.1002/cm.21279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/26/2016] [Accepted: 01/26/2016] [Indexed: 11/11/2022]
Abstract
Cells are known to respond over time to mechanical stimuli, even actively generating force at longer times. In this paper, a microstructural filament-based cytoskeletal network model is extended to incorporate this active response, and a computational study to assess the influence on relaxation behaviour was performed. The incorporation of an active response was achieved by including a strain energy function of contractile activity from the cross-linked actin filaments. A four-state chemical model and strain energy function was adopted, and generalisation to three dimensions and the macroscopic deformation field was performed by integration over the unit sphere. Computational results in MATLAB and ABAQUS/Explicit indicated an active cellular response over various time-scales, dependent on contractile parameters. Important features such as force generation and increasing cell stiffness due to prestress are qualitatively predicted. The work in this paper can easily be extended to encompass other filament-based cytoskeletal models as well.
Collapse
Affiliation(s)
- B Fallqvist
- Department of Solid Mechanics, Royal Institute of Technology, Teknikringen 8, 100 44 Stockholm, Sweden
| |
Collapse
|
238
|
Shenoy VB, Wang H, Wang X. A chemo-mechanical free-energy-based approach to model durotaxis and extracellular stiffness-dependent contraction and polarization of cells. Interface Focus 2016; 6:20150067. [PMID: 26855753 DOI: 10.1098/rsfs.2015.0067] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We propose a chemo-mechanical model based on stress-dependent recruitment of myosin motors to describe how the contractility, polarization and strain in cells vary with the stiffness of their surroundings and their shape. A contractility tensor, which depends on the distribution of myosin motors, is introduced to describe the chemical free energy of the cell due to myosin recruitment. We explicitly include the contributions to the free energy that arise from mechanosensitive signalling pathways (such as the SFX, Rho-Rock and MLCK pathways) through chemo-mechanical coupling parameters. Taking the variations of the total free energy, which consists of the chemical and mechanical components, in accordance with the second law of thermodynamics provides equations for the temporal evolution of the active stress and the contractility tensor. Following this approach, we are able to recover the well-known Hill relation for active stresses, based on the fundamental principles of irreversible thermodynamics rather than phenomenology. We have numerically implemented our free energy-based approach to model spatial distribution of strain and contractility in (i) cells supported by flexible microposts, (ii) cells on two-dimensional substrates, and (iii) cells in three-dimensional matrices. We demonstrate how the polarization of the cells and the orientation of stress fibres can be deduced from the eigenvalues and eigenvectors of the contractility tensor. Our calculations suggest that the chemical free energy of the cell decreases with the stiffness of the extracellular environment as the cytoskeleton polarizes in response to stress-dependent recruitment of molecular motors. The mechanical energy, which includes the strain energy and motor potential energy, however, increases with stiffness, but the overall energy is lower for cells in stiffer environments. This provides a thermodynamic basis for durotaxis, whereby cells preferentially migrate towards stiffer regions of the extracellular environment. Our models also explain, from an energetic perspective, why the shape of the cells can change in response to stiffness of the surroundings. The effect of the stiffness of the nucleus on its shape and the orientation of the stress fibres is also studied for all the above geometries. Along with making testable predictions, we have estimated the magnitudes of the chemo-mechanical coupling parameters for myofibroblasts based on data reported in the literature.
Collapse
Affiliation(s)
- Vivek B Shenoy
- Department of Materials Science and Engineering , University of Pennsylvania , Philadelphia, PA 19104 , USA
| | - Hailong Wang
- Department of Materials Science and Engineering , University of Pennsylvania , Philadelphia, PA 19104 , USA
| | - Xiao Wang
- Department of Materials Science and Engineering , University of Pennsylvania , Philadelphia, PA 19104 , USA
| |
Collapse
|
239
|
Fu PC, Tang RH, Wan Y, Xie MJ, Wang W, Luo X, Yu ZY. ROCK inhibition with fasudil promotes early functional recovery of spinal cord injury in rats by enhancing microglia phagocytosis. ACTA ACUST UNITED AC 2016; 36:31-36. [DOI: 10.1007/s11596-016-1537-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 10/13/2015] [Indexed: 02/06/2023]
|
240
|
Shi Y, Zhang L, Pu H, Mao L, Hu X, Jiang X, Xu N, Stetler RA, Zhang F, Liu X, Leak RK, Keep RF, Ji X, Chen J. Rapid endothelial cytoskeletal reorganization enables early blood-brain barrier disruption and long-term ischaemic reperfusion brain injury. Nat Commun 2016; 7:10523. [PMID: 26813496 PMCID: PMC4737895 DOI: 10.1038/ncomms10523] [Citation(s) in RCA: 321] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 12/22/2015] [Indexed: 12/26/2022] Open
Abstract
The mechanism and long-term consequences of early blood–brain barrier (BBB) disruption after cerebral ischaemic/reperfusion (I/R) injury are poorly understood. Here we discover that I/R induces subtle BBB leakage within 30–60 min, likely independent of gelatinase B/MMP-9 activities. The early BBB disruption is caused by the activation of ROCK/MLC signalling, persistent actin polymerization and the disassembly of junctional proteins within microvascular endothelial cells (ECs). Furthermore, the EC alterations facilitate subsequent infiltration of peripheral immune cells, including MMP-9-producing neutrophils/macrophages, resulting in late-onset, irreversible BBB damage. Inactivation of actin depolymerizing factor (ADF) causes sustained actin polymerization in ECs, whereas EC-targeted overexpression of constitutively active mutant ADF reduces actin polymerization and junctional protein disassembly, attenuates both early- and late-onset BBB impairment, and improves long-term histological and neurological outcomes. Thus, we identify a previously unexplored role for early BBB disruption in stroke outcomes, whereby BBB rupture may be a cause rather than a consequence of parenchymal cell injury. Matrix metalloproteinases (MMPs) released from infiltrating immune cells are a major contributor to blood-brain barrier (BBB) breakdown following stroke. Here, the authors identify an early, MMP-independent BBB breakdown mechanism caused by rapid cytoskeletal rearrangements in endothelial cells, which could be inhibited by ADF.
Collapse
Affiliation(s)
- Yejie Shi
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.,China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Lili Zhang
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.,State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.,Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania 15261, USA
| | - Hongjian Pu
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.,State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Leilei Mao
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.,State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Xiaoming Hu
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.,State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.,Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania 15261, USA
| | - Xiaoyan Jiang
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.,State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Na Xu
- State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - R Anne Stetler
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.,State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.,Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania 15261, USA
| | - Feng Zhang
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.,State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.,Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania 15261, USA
| | - Xiangrong Liu
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.,China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania 15282, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Xunming Ji
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jun Chen
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.,State Key Laboratory of Medical Neurobiology, Institute of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.,Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania 15261, USA
| |
Collapse
|
241
|
Rho-Associated Kinase Inhibitors Promote Microglial Uptake Via the ERK Signaling Pathway. Neurosci Bull 2016; 32:83-91. [PMID: 26779919 DOI: 10.1007/s12264-016-0013-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 11/26/2015] [Indexed: 12/17/2022] Open
Abstract
Microglia are immunocompetent cells in the central nervous system that take up tissue debris and pathogens. Rho-associated kinase (ROCK) has been identified as an important regulator of uptake, proliferation, secretion, and differentiation in a number of cell types. Although ROCK plays critical roles in the microglial secretion of inflammatory factors, migration, and morphology, its effects on microglial uptake activity have not been well characterized. In the present study, we found that treatment of BV2 microglia and primary microglia with the ROCK inhibitors Y27632 and fasudil increased uptake activity and was associated with morphological changes. Furthermore, western blots showed that this increase in uptake activity was mediated through the extracellular-signal-regulated kinase (ERK) signaling cascade, indicating the importance of ROCK in regulating microglial uptake activity.
Collapse
|
242
|
Lehtimäki J, Hakala M, Lappalainen P. Actin Filament Structures in Migrating Cells. Handb Exp Pharmacol 2016; 235:123-152. [PMID: 27469496 DOI: 10.1007/164_2016_28] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell migration is necessary for several developmental processes in multicellular organisms. Furthermore, many physiological processes such as wound healing and immunological events in adult animals are dependent on cell migration. Consequently, defects in cell migration are linked to various diseases including immunological disorders as well as cancer progression and metastasis formation. Cell migration is driven by specific protrusive and contractile actin filament structures, but the types and relative contributions of these actin filament arrays vary depending on the cell type and the environment of the cell. In this chapter, we introduce the most important actin filament structures that contribute to mesenchymal and amoeboid cell migration modes and discuss the mechanisms by which the assembly and turnover of these structures are controlled by various actin-binding proteins.
Collapse
Affiliation(s)
- Jaakko Lehtimäki
- Institute of Biotechnology, University of Helsinki, 56, 00014, Helsinki, Finland
| | - Markku Hakala
- Institute of Biotechnology, University of Helsinki, 56, 00014, Helsinki, Finland
| | - Pekka Lappalainen
- Institute of Biotechnology, University of Helsinki, 56, 00014, Helsinki, Finland.
| |
Collapse
|
243
|
Fallqvist B, Fielden ML, Pettersson T, Nordgren N, Kroon M, Gad AKB. Experimental and computational assessment of F-actin influence in regulating cellular stiffness and relaxation behaviour of fibroblasts. J Mech Behav Biomed Mater 2015; 59:168-184. [PMID: 26766328 DOI: 10.1016/j.jmbbm.2015.11.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Revised: 11/27/2015] [Accepted: 11/30/2015] [Indexed: 12/28/2022]
Abstract
In biomechanics, a complete understanding of the structures and mechanisms that regulate cellular stiffness at a molecular level remain elusive. In this paper, we have elucidated the role of filamentous actin (F-actin) in regulating elastic and viscous properties of the cytoplasm and the nucleus. Specifically, we performed colloidal-probe atomic force microscopy (AFM) on BjhTERT fibroblast cells incubated with Latrunculin B (LatB), which results in depolymerisation of F-actin, or DMSO control. We found that the treatment with LatB not only reduced cellular stiffness, but also greatly increased the relaxation rate for the cytoplasm in the peripheral region and in the vicinity of the nucleus. We thus conclude that F-actin is a major determinant in not only providing elastic stiffness to the cell, but also in regulating its viscous behaviour. To further investigate the interdependence of different cytoskeletal networks and cell shape, we provided a computational model in a finite element framework. The computational model is based on a split strain energy function of separate cellular constituents, here assumed to be cytoskeletal components, for which a composite strain energy function was defined. We found a significant influence of cell geometry on the predicted mechanical response. Importantly, the relaxation behaviour of the cell can be characterised by a material model with two time constants that have previously been found to predict mechanical behaviour of actin and intermediate filament networks. By merely tuning two effective stiffness parameters, the model predicts experimental results in cells with a partly depolymerised actin cytoskeleton as well as in untreated control. This indicates that actin and intermediate filament networks are instrumental in providing elastic stiffness in response to applied forces, as well as governing the relaxation behaviour over shorter and longer time-scales, respectively.
Collapse
Affiliation(s)
- Björn Fallqvist
- Department of Solid Mechanics, KTH Royal Institute of Technology, Teknikringen 8, 100 44 Stockholm, Sweden.
| | - Matthew L Fielden
- NANOLAB, KTH Royal Institute of Technology, Roslagstullsbacken 21, 100 44 Stockholm, Sweden.
| | - Torbjörn Pettersson
- Fibre and Polymer Technology, KTH Royal Institute of Technology, Teknikringen 56-58, 100 44 Stockholm, Sweden.
| | - Niklas Nordgren
- SP Chemistry, Materials and Surfaces, SP Technical Research Institute of Sweden, 114 86 Stockholm, Sweden.
| | - Martin Kroon
- Department of Solid Mechanics, KTH Royal Institute of Technology, Teknikringen 8, 100 44 Stockholm, Sweden.
| | - Annica K B Gad
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Nobels väg 16, 171 77 Stockholm, Sweden.
| |
Collapse
|
244
|
Nalluri SM, O'Connor JW, Gomez EW. Cytoskeletal signaling in TGFβ-induced epithelial-mesenchymal transition. Cytoskeleton (Hoboken) 2015; 72:557-69. [PMID: 26543012 DOI: 10.1002/cm.21263] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/04/2015] [Accepted: 11/04/2015] [Indexed: 12/13/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a physiological process that plays an important role in embryonic development and wound healing and is appropriated during pathological conditions including fibrosis and cancer metastasis. EMT can be initiated by a variety of factors, including transforming growth factor (TGF)-β, and is characterized by loss of epithelial features including cell-cell contacts and apicobasal polarity and acquisition of a motile, mesenchymal phenotype. A key feature of EMT is reorganization of the cytoskeleton and recent studies have elucidated regulation mechanisms governing this process. This review describes changes in gene expression patterns of cytoskeletal associated proteins during TGFβ-induced EMT. It further reports TGFβ-induced intracellular signaling cascades that regulate cytoskeletal reorganization during EMT. Finally, it highlights how changes in cytoskeletal architecture during EMT can regulate gene expression, thus further promoting EMT progression.
Collapse
Affiliation(s)
- Sandeep M Nalluri
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, 16802
| | - Joseph W O'Connor
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, 16802
| | - Esther W Gomez
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, 16802.,Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, 16802
| |
Collapse
|
245
|
Puthumana J, Jose S, Philip R, Singh ISB. Cellular and molecular markers in monitoring the fate of lymphoid cell culture from Penaeus monodon Fabricius (1798). FISH & SHELLFISH IMMUNOLOGY 2015; 47:893-901. [PMID: 26052014 DOI: 10.1016/j.fsi.2015.05.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 05/25/2015] [Accepted: 05/28/2015] [Indexed: 06/04/2023]
Abstract
Lymphoid cell culture from penaeid shrimps has gained much acceptance as an in vitro platform to facilitate research on the development of prophylaxis, and therapeutic strategies against viruses and for cell line development. However, lymphoid cells can be used as platform for in vitro research, only if they are in metabolically and mitotically active state in vitro with unaltered cell surface receptors. Through this study, we addressed the response of lymphoid cells to a new microenvironment at cellular and molecular levels; including the study of mitotic events, DNA synthesis, expression profile of cell cycle genes, cytoskeleton organization, metabolic activity and viral susceptibility. The S-phase entry and synthesis of new DNA was recorded by immunoflourescent technique. Cdc2, CycA, CycB, EF-1α and BUB3 genes involved in cell cycle were studied in both the cells and tissue, of which EF-1α showed an elevated expression in cells in vitro (∼ 19.7%). Cytoskeleton network of the cell was examined by studying the organization of actin filaments. As the markers for metabolic status, mitochondrial dehydrogenase, protein synthesis and glucose assimilation by the cells were also assessed. Viral susceptibility of the cell was determined using WSSV to confirm the preservation of cellular receptors. This study envisages to strengthen the shrimp cell line research and to bring forth lymphoid cell culture system as a 'model' in vitro system for shrimp and crustaceans altogether.
Collapse
Affiliation(s)
- Jayesh Puthumana
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala 682016, India; Department of Marine Biology, Microbiology and Biochemistry, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala 682016, India.
| | - Seena Jose
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala 682016, India
| | - Rosamma Philip
- Department of Marine Biology, Microbiology and Biochemistry, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala 682016, India
| | - I S Bright Singh
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala 682016, India.
| |
Collapse
|
246
|
Li N, Tang EI, Cheng CY. Regulation of blood-testis barrier by actin binding proteins and protein kinases. Reproduction 2015; 151:R29-41. [PMID: 26628556 DOI: 10.1530/rep-15-0463] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/01/2015] [Indexed: 12/13/2022]
Abstract
The blood-testis barrier (BTB) is an important ultrastructure in the testis, since the onset of meiosis and spermiogenesis coincides with the establishment of a functional barrier in rodents and humans. It is also noted that a delay in the assembly of a functional BTB following treatment of neonatal rats with drugs such as diethylstilbestrol or adjudin also delays the first wave of spermiation. While the BTB is one of the tightest blood-tissue barriers, it undergoes extensive remodeling, in particular, at stage VIII of the epithelial cycle to facilitate the transport of preleptotene spermatocytes connected in clones across the immunological barrier. Without this timely transport of preleptotene spermatocytes derived from type B spermatogonia, meiosis will be arrested, causing aspermatogenesis. Yet the biology and regulation of the BTB remains largely unexplored since the morphological studies in the 1970s. Recent studies, however, have shed new light on the biology of the BTB. Herein, we critically evaluate some of these findings, illustrating that the Sertoli cell BTB is regulated by actin-binding proteins (ABPs), likely supported by non-receptor protein kinases, to modulate the organization of actin microfilament bundles at the site. Furthermore, microtubule-based cytoskeleton is also working in concert with the actin-based cytoskeleton to confer BTB dynamics. This timely review provides an update on the unique biology and regulation of the BTB based on the latest findings in the field, focusing on the role of ABPs and non-receptor protein kinases.
Collapse
Affiliation(s)
- Nan Li
- The Mary M. Wohlford Laboratory for Male Contraceptive ResearchCenter for Biomedical Research, Population Council, 1230 York Avenue, New York, New York 10065, USA
| | - Elizabeth I Tang
- The Mary M. Wohlford Laboratory for Male Contraceptive ResearchCenter for Biomedical Research, Population Council, 1230 York Avenue, New York, New York 10065, USA
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive ResearchCenter for Biomedical Research, Population Council, 1230 York Avenue, New York, New York 10065, USA
| |
Collapse
|
247
|
Branchfield K, Li R, Lungova V, Verheyden JM, McCulley D, Sun X. A three-dimensional study of alveologenesis in mouse lung. Dev Biol 2015; 409:429-41. [PMID: 26632490 DOI: 10.1016/j.ydbio.2015.11.017] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 11/23/2015] [Accepted: 11/23/2015] [Indexed: 01/08/2023]
Abstract
Alveologenesis is the final step of lung maturation, which subdivides the alveolar region of the lung into smaller units called alveoli. Each of the nascent dividers serves as a new gas-exchange surface, and collectively they drastically increase the surface area for breathing. Disruption of alveologenesis results in simplification of alveoli, as is seen in premature infants diagnosed with bronchopulmonary dysplasia (BPD), a prevalent lung disease that is often associated with lifelong breathing deficiencies. To date, a majority of studies of alveologenesis rely on two-dimensional (2D) analysis of tissue sections. Given that an overarching theme of alveologenesis is thinning and extension of the epithelium and mesenchyme to facilitate gas exchange, often only a small portion of a cell or a cellular structure is represented in a single 2D plane. Here, we use a three-dimensional (3D) approach to examine the structural architecture and cellular composition of myofibroblasts, alveolar type 2 cells, elastin and lipid droplets in normal as well as BPD-like mouse lung. We found that 2D finger-like septal crests, commonly used to depict growing alveolar septae, are often artifacts of sectioning through fully established alveolar walls. Instead, a more accurate representation of growing septae are 3D ridges that are lined by platelet-derived growth factor receptor alpha (PDGFRA) and alpha smooth muscle actin (α-SMA)-expressing myofibroblasts, as well as the elastin fibers that they produce. Accordingly in 3D, both α-SMA and elastin were each found in connected networks underlying the 3D septal ridges rather than as isolated dots at the tip of 2D septal crests. Analysis through representative stages of alveologenesis revealed unappreciated dynamic changes in these patterns. PDGFRA-expressing cells are only α-SMA-positive during the first phase of alveologenesis, but not in the second phase, suggesting that the two phases of septae formation may be driven by distinct mechanisms. Thin elastin fibers are already present in the alveolar region prior to alveologenesis, suggesting that during alveologenesis, there is not only new elastin deposition, but also extensive remodeling to transform thin and uniformly distributed fibers into thick cables that rim the nascent septae. Analysis of several genetic as well as hyperoxia-induced models of BPD revealed that the myofibroblast organization is perturbed in all, regardless of whether the origin of defect is epithelial, mesenchymal, endothelial or environmental. Finally, analysis of relative position of PDGFRA-positive cells and alveolar type 2 cells reveal that during alveologenesis, these two cell types are not always adjacent to one another. This result suggests that the niche and progenitor relationship afforded by their close juxtaposition in the adult lung may be a later acquired property. These insights revealed by 3D reconstruction of the septae set the foundation for future investigations of the mechanisms driving normal alveologenesis, as well as causes of alveolar simplification in BPD.
Collapse
Affiliation(s)
- Kelsey Branchfield
- Laboratory of Genetics, University of Wisconsin-Madison Madison, WI 52706, United States
| | - Rongbo Li
- Laboratory of Genetics, University of Wisconsin-Madison Madison, WI 52706, United States
| | - Vlasta Lungova
- Department of Surgery, University of Wisconsin-Madison Madison, WI 53706, United States
| | - Jamie M Verheyden
- Laboratory of Genetics, University of Wisconsin-Madison Madison, WI 52706, United States
| | - David McCulley
- Department of Pediatrics University of Wisconsin-Madison Madison, WI 53706, United States
| | - Xin Sun
- Laboratory of Genetics, University of Wisconsin-Madison Madison, WI 52706, United States.
| |
Collapse
|
248
|
Rho/Rock signal transduction pathway is required for MSC tenogenic differentiation. Bone Res 2015; 3:15015. [PMID: 26509098 PMCID: PMC4605238 DOI: 10.1038/boneres.2015.15] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 03/20/2015] [Accepted: 04/20/2015] [Indexed: 01/20/2023] Open
Abstract
Mesenchymal stem cell (MSC)-based treatments have shown promise for improving tendon healing and repair. MSCs have the potential to differentiate into multiple lineages in response to select chemical and physical stimuli, including into tenocytes. Cell elongation and cytoskeletal tension have been shown to be instrumental to the process of MSC differentiation. Previous studies have shown that inhibition of stress fiber formation leads MSCs to default toward an adipogenic lineage, which suggests that stress fibers are required for MSCs to sense the environmental factors that can induce differentiation into tenocytes. As the Rho/ROCK signal transduction pathway plays a critical role in both stress fiber formation and in cell sensation, we examined whether the activation of this pathway was required when inducing MSC tendon differentiation using rope-like silk scaffolds. To accomplish this, we employed a loss-of-function approach by knocking out ROCK, actin and myosin (two other components of the pathway) using the specific inhibitors Y-27632, Latrunculin A and blebbistatin, respectively. We demonstrated that independently disrupting the cytoskeleton and the Rho/ROCK pathway abolished the expression of tendon differentiation markers and led to a loss of spindle morphology. Together, these studies suggest that the tension that is generated by MSC elongation is essential for MSC teno-differentiation and that the Rho/ROCK pathway is a critical mediator of tendon differentiation on rope-like silk scaffolds.
Collapse
|
249
|
Abstract
Cellular processes, including morphogenesis, polarization, and motility, rely on a variety of actin-based structures. Although the biochemical composition and filament organization of these structures are different, they often emerge from a common origin. This is possible because the actin structures are highly dynamic. Indeed, they assemble, grow, and disassemble in a time scale of a second to a minute. Therefore, the reorganization of a given actin structure can promote the formation of another. Here, we discuss such transitions and illustrate them with computer simulations.
Collapse
Affiliation(s)
- Gaëlle Letort
- Laboratoire de Physiologie Cellulaire et Végétale, Institut de Recherches en Technologies et Sciences pour le Vivant, iRTSV, CNRS/CEA/UGA, Grenoble, France
| | - Hajer Ennomani
- Laboratoire de Physiologie Cellulaire et Végétale, Institut de Recherches en Technologies et Sciences pour le Vivant, iRTSV, CNRS/CEA/UGA, Grenoble, France
| | - Laurène Gressin
- Laboratoire de Physiologie Cellulaire et Végétale, Institut de Recherches en Technologies et Sciences pour le Vivant, iRTSV, CNRS/CEA/UGA, Grenoble, France
| | - Manuel Théry
- Laboratoire de Physiologie Cellulaire et Végétale, Institut de Recherches en Technologies et Sciences pour le Vivant, iRTSV, CNRS/CEA/UGA, Grenoble, France.,Unité de Thérapie Cellulaire, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, INSERM/AP-HP/Université Paris Diderot, Paris, France
| | - Laurent Blanchoin
- Laboratoire de Physiologie Cellulaire et Végétale, Institut de Recherches en Technologies et Sciences pour le Vivant, iRTSV, CNRS/CEA/UGA, Grenoble, France
| |
Collapse
|
250
|
Ach T, Tolstik E, Messinger JD, Zarubina AV, Heintzmann R, Curcio CA. Lipofuscin redistribution and loss accompanied by cytoskeletal stress in retinal pigment epithelium of eyes with age-related macular degeneration. Invest Ophthalmol Vis Sci 2015; 56:3242-52. [PMID: 25758814 DOI: 10.1167/iovs.14-16274] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Lipofuscin (LF) and melanolipofuscin (MLF) of the retinal pigment epithelium (RPE) are the principal sources of autofluorescence (AF) signals in clinical fundus-AF imaging. Few details about the subcellular distribution of AF organelles in AMD are available. We describe the impact of aging and AMD on RPE morphology revealed by the distribution of AF LF/MLF granules and actin cytoskeleton in human tissues. METHODS Thirty-five RPE-Bruch's membrane flatmounts from 35 donors were prepared (postmortem: ≤4 hours). Ex vivo fundus examination at the time of accession revealed either absence of chorioretinal pathologies (10 tissues; mean age: 83.0 ± 2.6 years) or stages of AMD (25 tissues; 85.0 ± 5.8 years): early AMD, geographic atrophy, and late exudative AMD. Retinal pigment epithelium cytoskeleton was labeled with AlexaFluor647-Phalloidin. Tissues were imaged on a spinning-disk fluorescence microscope and a high-resolution structured illumination microscope. RESULTS Age-related macular degeneration impacts individual RPE cells by (1) lipofuscin redistribution by (i) degranulation (granule-by-granule loss) and/or (ii) aggregation and apparent shedding into the extracellular space; (2) enlarged RPE cell area and conversion from convex to irregular and sometimes concave polygons; and (3) cytoskeleton derangement including separations and breaks around subretinal deposits, thickening, and stress fibers. CONCLUSIONS We report an extensive and systematic en face analysis of LF/MLF-AF in AMD eyes. Redistribution and loss of AF granules are among the earliest AMD changes and could reduce fundus AF signal attributable to RPE at these locations. Data can enhance the interpretation of clinical fundus-AF and provide a basis for future quantitative studies.
Collapse
Affiliation(s)
- Thomas Ach
- University of Alabama at Birmingham Department of Ophthalmology, Birmingham, Alabama, United States 2University Hospital Würzburg, Department of Ophthalmology, Würzburg, Germany
| | - Elen Tolstik
- Leibniz Institute of Photonic Technology, Jena, Germany 5King's College London, Randall Division of Cell & Molecular Biophysics, London, United Kingdom
| | - Jeffrey D Messinger
- University of Alabama at Birmingham Department of Ophthalmology, Birmingham, Alabama, United States
| | - Anna V Zarubina
- University of Alabama at Birmingham Department of Ophthalmology, Birmingham, Alabama, United States
| | - Rainer Heintzmann
- Leibniz Institute of Photonic Technology, Jena, Germany 5King's College London, Randall Division of Cell & Molecular Biophysics, London, United Kingdom
| | - Christine A Curcio
- University of Alabama at Birmingham Department of Ophthalmology, Birmingham, Alabama, United States
| |
Collapse
|