51
|
Kahue CN, Jerrell RJ, Parekh A. Expression of human papillomavirus oncoproteins E6 and E7 inhibits invadopodia activity but promotes cell migration in HPV-positive head and neck squamous cell carcinoma cells. Cancer Rep (Hoboken) 2018; 1:e1125. [PMID: 32721084 PMCID: PMC7941430 DOI: 10.1002/cnr2.1125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 12/30/2022] Open
Abstract
Background The rapid increase in the incidence of head and neck squamous cell carcinoma (HNSCC) is caused by high‐risk human papillomavirus (HPV) infections. The HPV oncogenes E6 and E7 promote carcinogenesis by disrupting signaling pathways that control survival and proliferation. Although these cancers are often diagnosed with metastases, the mechanisms that regulate their dissemination are unknown. Aims The aim of this study was to determine whether the HPV‐16 E6 and E7 oncogenes affected the invasive and migratory properties of HNSCC cells which promote their spread and metastasis. Methods and results Invasiveness was determined using invadopodia assays which allow for quantitation of extracellular matrix (ECM) degradation by invadopodia which are proteolytic membrane protrusions that facilitate invasion. Using cell lines and genetic manipulations, we found that HPV inhibited invadopodia activity in aggressive cell lines which was mediated by the E6 and E7 oncogenes. Given these findings, we also tested whether HPV caused differences in the migratory ability of HNSCC cells using Transwell assays. In contrast to our invadopodia results, we found no correlation between HPV status and cell migration; however, blocking the expression of the E6 and E7 oncoproteins in a HPV‐positive (HPV+) HNSCC cell line resulted in decreased migration. Conclusions Our data suggest that the E6 and E7 oncoproteins are negative regulators of invadopodia activity but may promote migration in HPV+ HNSCC cells. Despite the need for ECM proteolysis to penetrate most tissues, the unique structure of the head and neck tissues in which these cancers arise may facilitate the spread of migratory cancer cells without significant proteolytic ability.
Collapse
Affiliation(s)
- Charissa N Kahue
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel J Jerrell
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Aron Parekh
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
52
|
Abstract
The Hippo signaling pathway controls nuclear accumulation and stability of the transcriptional coregulator YAP and its paralog TAZ. The activity of Hippo-YAP signaling is influenced not only by biochemical signals, but also by cell shape and mechanical tension transmitted through cell-cell junctions and cell-matrix adhesions. Data accumulated thus far indicates that the actin cytoskeleton is a key mediator of the regulation of Hippo-YAP signaling by means of a variety of biochemical and mechanical cues. In this review, we have outlined the role of actin dynamics and actin-associated proteins in the regulation of Hippo-YAP signaling. In addition, we discuss actinmediated regulation of YAP/TAZ activity independent of the core Hippo kinases MST and LATS. Although our understanding of the link between Hippo-YAP signaling and the actin cytoskeleton is progressing rapidly, many open questions remain. [BMB Reports 2018; 51(3): 151-156].
Collapse
Affiliation(s)
- Jimyung Seo
- Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Korea
| | - Joon Kim
- Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Korea
| |
Collapse
|
53
|
Yap AS, Duszyc K, Viasnoff V. Mechanosensing and Mechanotransduction at Cell-Cell Junctions. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028761. [PMID: 28778874 DOI: 10.1101/cshperspect.a028761] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cell adhesion systems are defined by their ability to resist detachment force. Our understanding of the biology of cell-cell adhesions has recently been transformed by the realization that many of the forces that act on those adhesions are generated by the cells that they couple together; and that force at adhesive junctions can be sensed to regulate cell behavior. Here, we consider the mechanisms responsible for applying force to cell-cell junctions and the mechanosensory pathways that detect those forces. We focus on cadherins, as these are the best-studied examples to date, but it is likely that similar principles will apply to other molecular systems that can engage with force-generators within cells and physically couple those cells together.
Collapse
Affiliation(s)
- Alpha S Yap
- Institute for Molecular Bioscience, Division of Cell Biology and Molecular Medicine, The University of Queensland, St. Lucia, Brisbane 4072, Australia
| | - Kinga Duszyc
- Institute for Molecular Bioscience, Division of Cell Biology and Molecular Medicine, The University of Queensland, St. Lucia, Brisbane 4072, Australia
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, Singapore 117411.,CNRS, Singapore 117411
| |
Collapse
|
54
|
TRPM7 controls mesenchymal features of breast cancer cells by tensional regulation of SOX4. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2409-2419. [DOI: 10.1016/j.bbadis.2018.04.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/13/2018] [Accepted: 04/18/2018] [Indexed: 01/04/2023]
|
55
|
McKenzie AJ, Hicks SR, Svec KV, Naughton H, Edmunds ZL, Howe AK. The mechanical microenvironment regulates ovarian cancer cell morphology, migration, and spheroid disaggregation. Sci Rep 2018; 8:7228. [PMID: 29740072 PMCID: PMC5940803 DOI: 10.1038/s41598-018-25589-0] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/24/2018] [Indexed: 01/13/2023] Open
Abstract
There is growing appreciation of the importance of the mechanical properties of the tumor microenvironment on disease progression. However, the role of extracellular matrix (ECM) stiffness and cellular mechanotransduction in epithelial ovarian cancer (EOC) is largely unknown. Here, we investigated the effect of substrate rigidity on various aspects of SKOV3 human EOC cell morphology and migration. Young’s modulus values of normal mouse peritoneum, a principal target tissue for EOC metastasis, were determined by atomic force microscopy (AFM) and hydrogels were fabricated to mimic these values. We find that cell spreading, focal adhesion formation, myosin light chain phosphorylation, and cellular traction forces all increase on stiffer matrices. Substrate rigidity also positively regulates random cell migration and, importantly, directional increases in matrix tension promote SKOV3 cell durotaxis. Matrix rigidity also promotes nuclear translocation of YAP1, an oncogenic transcription factor associated with aggressive metastatic EOC. Furthermore, disaggregation of multicellular EOC spheroids, a behavior associated with dissemination and metastasis, is enhanced by matrix stiffness through a mechanotransduction pathway involving ROCK, actomyosin contractility, and FAK. Finally, this pattern of mechanosensitivity is maintained in highly metastatic SKOV3ip.1 cells. These results establish that the mechanical properties of the tumor microenvironment may play a role in EOC metastasis.
Collapse
Affiliation(s)
- Andrew J McKenzie
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Stephanie R Hicks
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Kathryn V Svec
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Hannah Naughton
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Zöe L Edmunds
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States
| | - Alan K Howe
- University of Vermont Larner College of Medicine, Department of Pharmacology, and the University of Vermont Cancer Center, Burlington, United States.
| |
Collapse
|
56
|
Li S, Xiong N, Peng Y, Tang K, Bai H, Lv X, Jiang Y, Qin X, Yang H, Wu C, Zhou P, Liu Y. Acidic pHe regulates cytoskeletal dynamics through conformational integrin β1 activation and promotes membrane protrusion. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2395-2408. [PMID: 29698684 DOI: 10.1016/j.bbadis.2018.04.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/02/2018] [Accepted: 04/19/2018] [Indexed: 12/15/2022]
Abstract
An acidic extracellular pH (pHe) in the tumor microenvironment has been suggested to facilitate tumor growth and metastasis. However, the molecular mechanisms by which tumor cells sense acidic signal to induce a transition to an aggressive phenotype remain elusive. Here, we showed that an acidic pHe (pH 6.5) stimulation resulted in protrusion and epithelial-mesenchymal transition (EMT) of cancer cells, which promoted migration and matrix degeneration. Using computational molecular dynamics simulations, we reported acidic pHe-induced opening of the Integrin dimers (α5β1) headpiece which indicated the activation of integrin. Moreover, acidic pHe promoted maturation of focal adhesions, temporal activation of Rho GTPases and microfilament reorganization through integrin β1-activated FAK signaling. Furthermore, mechanical balance of cytoskeleton (actin, tubulin and vimentin) contributed to acidic pHe-triggered protrusion and morphology change. Taken together, these findings revealed that integrin β1 could be a novel pH-regulated sensitive molecule which confers protrusion and malignant phenotype of cancer cells.
Collapse
Affiliation(s)
- Shun Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Niya Xiong
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Yueting Peng
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Kai Tang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Hongxia Bai
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Xiaoying Lv
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Ying Jiang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Xiang Qin
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Hong Yang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Peng Zhou
- Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China.
| |
Collapse
|
57
|
Kelley CA, Wirshing ACE, Zaidel-Bar R, Cram EJ. The myosin light-chain kinase MLCK-1 relocalizes during Caenorhabditis elegans ovulation to promote actomyosin bundle assembly and drive contraction. Mol Biol Cell 2018; 29:1975-1991. [PMID: 30088798 PMCID: PMC6232974 DOI: 10.1091/mbc.e18-01-0056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
We identify the Caenorhabditis elegans myosin light-chain kinase, MLCK-1, required for contraction of spermathecae. During contraction, MLCK-1 moves from the apical cell boundaries to the basal actomyosin bundles, where it stabilizes myosin downstream of calcium signaling. MLCK and ROCK act in distinct subsets of cells to coordinate the timing of contraction.
Collapse
Affiliation(s)
| | | | - Ronen Zaidel-Bar
- Department of Cell and Developmental Biology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Erin J Cram
- Department of Biology, Northeastern University, Boston, MA 02115
| |
Collapse
|
58
|
The WAVE Regulatory Complex and Branched F-Actin Counterbalance Contractile Force to Control Cell Shape and Packing in the Drosophila Eye. Dev Cell 2018; 44:471-483.e4. [PMID: 29396116 DOI: 10.1016/j.devcel.2017.12.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 09/14/2017] [Accepted: 12/26/2017] [Indexed: 12/27/2022]
Abstract
Contractile forces eliminate cell contacts in many morphogenetic processes. However, mechanisms that balance contractile forces to promote subtler remodeling remain unknown. To address this gap, we investigated remodeling of Drosophila eye lattice cells (LCs), which preserve cell contacts as they narrow to form the edges of a multicellular hexagonal lattice. We found that during narrowing, LC-LC contacts dynamically constrict and expand. Similar to other systems, actomyosin-based contractile forces promote pulses of constriction. Conversely, we found that WAVE-dependent branched F-actin accumulates at LC-LC contacts during expansion and functions to expand the cell apical area, promote shape changes, and prevent elimination of LC-LC contacts. Finally, we found that small Rho GTPases regulate the balance of contractile and protrusive dynamics. These data suggest a mechanism by which WAVE regulatory complex-based F-actin dynamics antagonize contractile forces to regulate cell shape and tissue topology during remodeling and thus contribute to the robustness and precision of the process.
Collapse
|
59
|
Abstract
The actin cytoskeleton-a collection of actin filaments with their accessory and regulatory proteins-is the primary force-generating machinery in the cell. It can produce pushing (protrusive) forces through coordinated polymerization of multiple actin filaments or pulling (contractile) forces through sliding actin filaments along bipolar filaments of myosin II. Both force types are particularly important for whole-cell migration, but they also define and change the cell shape and mechanical properties of the cell surface, drive the intracellular motility and morphogenesis of membrane organelles, and allow cells to form adhesions with each other and with the extracellular matrix.
Collapse
Affiliation(s)
- Tatyana Svitkina
- Department of Biology, University of Pennsylvania, 221 Leidy Labs, Philadelphia, Pennsylvania 19104
| |
Collapse
|
60
|
Fu PC, Tang RH, Yu ZY, Xie MJ, Wang W, Luo X. The Rho-associated kinase inhibitors Y27632 and fasudil promote microglial migration in the spinal cord via the ERK signaling pathway. Neural Regen Res 2018; 13:677-683. [PMID: 29722320 PMCID: PMC5950678 DOI: 10.4103/1673-5374.230294] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Rho-associated kinase (ROCK) is a key regulatory protein involved in inflammatory secretion in microglia in the central nervous system. Our previous studies showed that ROCK inhibition enhances phagocytic activity in microglia through the extracellular signal-regulated kinase (ERK) signaling pathway, but its effect on microglial migration was unknown. Therefore, in this study, we investigated the effects of the ROCK inhibitors Y27632 and fasudil on the migratory activity of primary cultured microglia isolated from the spinal cord, and we examined the underlying mechanisms. The microglia were treated with Y27632, fasudil and/or the ERK inhibitor U0126. Cellular morphology was observed by immunofluorescence. Transwell chambers were used to assess cell migration. ERK levels were measured by in-cell western blot assay. Y27632 and fasudil increased microglial migration, and the microglia were irregularly shaped and had many small processes. These inhibitors also upregulated the levels of phosphorylated ERK protein. The ERK inhibitor U0126 suppressed these effects of Y27632 and fasudil. These findings suggest that the ROCK inhibitors Y27632 and fasudil promote microglial migration in the spinal cord through the ERK signaling pathway.
Collapse
Affiliation(s)
- Pei-Cai Fu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Rong-Hua Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhi-Yuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Neurological Diseases (Huazhong University of Science and Technology), Ministry of Education of China, Wuhan, Hubei Province, China
| | - Min-Jie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Neurological Diseases (Huazhong University of Science and Technology), Ministry of Education of China, Wuhan, Hubei Province, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Neurological Diseases (Huazhong University of Science and Technology), Ministry of Education of China, Wuhan, Hubei Province, China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
61
|
Sonavane PR, Wang C, Dzamba B, Weber GF, Periasamy A, DeSimone DW. Mechanical and signaling roles for keratin intermediate filaments in the assembly and morphogenesis of Xenopus mesendoderm tissue at gastrulation. Development 2017; 144:4363-4376. [PMID: 28982683 PMCID: PMC5769636 DOI: 10.1242/dev.155200] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 09/25/2017] [Indexed: 12/21/2022]
Abstract
The coordination of individual cell behaviors is a crucial step in the assembly and morphogenesis of tissues. Xenopus mesendoderm cells migrate collectively along a fibronectin (FN) substrate at gastrulation, but how the adhesive and mechanical forces required for these movements are generated and transmitted is unclear. Traction force microscopy (TFM) was used to establish that traction stresses are limited primarily to leading edge cells in mesendoderm explants, and that these forces are balanced by intercellular stresses in follower rows. This is further reflected in the morphology of these cells, with broad lamellipodial protrusions, mature focal adhesions and a gradient of activated Rac1 evident at the leading edge, while small protrusions, rapid turnover of immature focal adhesions and lack of a Rac1 activity gradient characterize cells in following rows. Depletion of keratin (krt8) with antisense morpholinos results in high traction stresses in follower row cells, misdirected protrusions and the formation of actin stress fibers anchored in streak-like focal adhesions. We propose that maintenance of mechanical integrity in the mesendoderm by keratin intermediate filaments is required to balance stresses within the tissue to regulate collective cell movements.
Collapse
Affiliation(s)
- Pooja R Sonavane
- Department of Cell Biology, School of Medicine, University of Virginia Health System, P.O. Box 800732, Charlottesville, VA 22908, USA
| | - Chong Wang
- Department of Cell Biology, School of Medicine, University of Virginia Health System, P.O. Box 800732, Charlottesville, VA 22908, USA
| | - Bette Dzamba
- Department of Cell Biology, School of Medicine, University of Virginia Health System, P.O. Box 800732, Charlottesville, VA 22908, USA
| | - Gregory F Weber
- Department of Cell Biology, School of Medicine, University of Virginia Health System, P.O. Box 800732, Charlottesville, VA 22908, USA
| | - Ammasi Periasamy
- Keck Center for Cellular Imaging, Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Douglas W DeSimone
- Department of Cell Biology, School of Medicine, University of Virginia Health System, P.O. Box 800732, Charlottesville, VA 22908, USA
| |
Collapse
|
62
|
Ishihara S, Inman DR, Li WJ, Ponik SM, Keely PJ. Mechano-Signal Transduction in Mesenchymal Stem Cells Induces Prosaposin Secretion to Drive the Proliferation of Breast Cancer Cells. Cancer Res 2017; 77:6179-6189. [PMID: 28972074 DOI: 10.1158/0008-5472.can-17-0569] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/27/2017] [Accepted: 09/25/2017] [Indexed: 01/15/2023]
Abstract
In response to chemical stimuli from cancer cells, mesenchymal stem cells (MSC) can differentiate into cancer-associated fibroblasts (CAF) and promote tumor progression. How mechanical stimuli such as stiffness of the extracellular matrix (ECM) contribute to MSC phenotype in cancer remains poorly understood. Here, we show that ECM stiffness leads to mechano-signal transduction in MSC, which promotes mammary tumor growth in part through secretion of the signaling protein prosaposin. On a stiff matrix, MSC cultured with conditioned media from mammary cancer cells expressed increased levels of α-smooth muscle actin, a marker of CAF, compared with MSC cultured on a soft matrix. By contrast, MSC cultured on a stiff matrix secreted prosaposin that promoted proliferation and survival of mammary carcinoma cells but inhibited metastasis. Our findings suggest that in addition to chemical stimuli, increased stiffness of the ECM in the tumor microenvironment induces differentiation of MSC to CAF, triggering enhanced proliferation and survival of mammary cancer cells. Cancer Res; 77(22); 6179-89. ©2017 AACR.
Collapse
Affiliation(s)
- Seiichiro Ishihara
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin.
| | - David R Inman
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Wan-Ju Li
- Departments of Orthopedics and Rehabilitation, and Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Patricia J Keely
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
63
|
Design of a nanocomposite substrate inducing adult stem cell assembly and progression toward an Epiblast-like or Primitive Endoderm-like phenotype via mechanotransduction. Biomaterials 2017; 144:211-229. [PMID: 28841465 DOI: 10.1016/j.biomaterials.2017.08.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/01/2017] [Accepted: 08/13/2017] [Indexed: 01/10/2023]
Abstract
This work shows that the active interaction between human umbilical cord matrix stem cells and Poly (l-lactide)acid (PLLA) and PLLA/Multi Walled Carbon Nanotubes (MWCNTs) nanocomposite films results in the stem cell assembly as a spheroid conformation and affects the stem cell fate transition. We demonstrated that spheroids directly respond to a tunable surface and the bulk properties (electric, dielectric and thermal) of plain and nanocomposite PLLA films by triggering a mechanotransduction axis. This stepwise process starts from tethering of the cells' focal adhesion proteins to the surface, together with the adherens junctions between cells. Both complexes transmit traction forces to F-Actin stress fibres that link Filamin-A and Myosin-IIA proteins, generating a biological scaffold, with increased stiffening conformation from PLLA to PLLA/MWCNTs, and enable the nucleoskeleton proteins to boost chromatin reprogramming processes. Herein, the opposite expression of NANOG and GATA6 transcription factors, together with other lineage specification related proteins, steer spheroids toward an Epiblast-like or Primitive Endoderm-like lineage commitment, depending on the absence or presence of 1 wt% MWCNTs, respectively. This work represents a pioneering effort to create a stem cell/material interface that can model the stem cell fate transition under growth culture conditions.
Collapse
|
64
|
Arora PD, Di Gregorio M, He P, McCulloch CA. TRPV4 mediates the Ca 2+ influx required for the interaction between flightless-1 and non-muscle myosin, and collagen remodeling. J Cell Sci 2017; 130:2196-2208. [PMID: 28526784 DOI: 10.1242/jcs.201665] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 05/14/2017] [Indexed: 12/15/2022] Open
Abstract
Fibroblasts remodel extracellular matrix collagen, in part, through phagocytosis. This process requires formation of cell extensions, which in turn involves interaction of the actin-binding protein flightless-1 (FliI) with non-muscle myosin IIA (NMMIIA; heavy chain encoded by MYH9) at cell-matrix adhesion sites. As Ca2+ plays a central role in controlling actomyosin-dependent functions, we examined how Ca2+ controls the generation of cell extensions and collagen remodeling. Ratio fluorimetry demonstrated localized Ca2+ influx at the extensions of fibroblasts. Western blotting and quantitative (q)PCR showed high expression levels of the Ca2+-permeable transient receptor potential vanilloid-4 (TRPV4) channel, which co-immunoprecipitated with β1 integrin and localized to adhesions. Treatment with α2β1-integrin-blocking antibody or the TRPV4-specific antagonist AB159908, as well as reduction of TRPV4 expression through means of siRNA, blocked Ca2+ influx. These treatments also inhibited the interaction of FliI with NMMIIA, reduced the number and length of cell extensions, and blocked collagen remodeling. Pulldown assays showed that Ca2+ depletion inhibited the interaction of purified FliI with NMMIIA filaments. Fluorescence resonance energy transfer experiments showed that FliI-NMMIIA interactions require Ca2+ influx. We conclude that Ca2+ influx through the TRPV4 channel regulates FliI-NMMIIA interaction, which in turn enables generation of the cell extensions essential for collagen remodeling.
Collapse
Affiliation(s)
- Pamma D Arora
- University of Toronto, Room 244, Fitzgerald Building, 150 College Street, Toronto, Ontario, M5S 3E2, Canada
| | - Madeleine Di Gregorio
- University of Toronto, Room 244, Fitzgerald Building, 150 College Street, Toronto, Ontario, M5S 3E2, Canada
| | - Pei He
- University of Toronto, Room 244, Fitzgerald Building, 150 College Street, Toronto, Ontario, M5S 3E2, Canada
| | - Christopher A McCulloch
- University of Toronto, Room 244, Fitzgerald Building, 150 College Street, Toronto, Ontario, M5S 3E2, Canada
| |
Collapse
|
65
|
Iamshanova O, Fiorio Pla A, Prevarskaya N. Molecular mechanisms of tumour invasion: regulation by calcium signals. J Physiol 2017; 595:3063-3075. [PMID: 28304082 DOI: 10.1113/jp272844] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 01/20/2017] [Indexed: 12/14/2022] Open
Abstract
Intracellular calcium (Ca2+ ) signals are key regulators of multiple cellular functions, both healthy and physiopathological. It is therefore unsurprising that several cancers present a strong Ca2+ homeostasis deregulation. Among the various hallmarks of cancer disease, a particular role is played by metastasis, which has a critical impact on cancer patients' outcome. Importantly, Ca2+ signalling has been reported to control multiple aspects of the adaptive metastatic cancer cell behaviour, including epithelial-mesenchymal transition, cell migration, local invasion and induction of angiogenesis (see Abstract Figure). In this context Ca2+ signalling is considered to be a substantial intracellular tool that regulates the dynamicity and complexity of the metastatic cascade. In the present study we review the spatial and temporal organization of Ca2+ fluxes, as well as the molecular mechanisms involved in metastasis, analysing the key steps which regulate initial tumour spread.
Collapse
Affiliation(s)
- Oksana Iamshanova
- Inserm U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Equipe Labellisée par la Ligue Nationale Contre le Cancer, SIRIC ONCOLille, University of Lille, 59656, Villeneuve d'Ascq, France
| | - Alessandra Fiorio Pla
- Inserm U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Equipe Labellisée par la Ligue Nationale Contre le Cancer, SIRIC ONCOLille, University of Lille, 59656, Villeneuve d'Ascq, France.,Department of Life Science and Systems Biology, University of Torino, Torino, Italy
| | - Natalia Prevarskaya
- Inserm U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Equipe Labellisée par la Ligue Nationale Contre le Cancer, SIRIC ONCOLille, University of Lille, 59656, Villeneuve d'Ascq, France
| |
Collapse
|
66
|
Chi Q, Shan J, Ding X, Yin T, Wang Y, Jia D, Wang G. Smart mechanosensing machineries enable migration of vascular smooth muscle cells in atherosclerosis-relevant 3D matrices. Cell Biol Int 2017; 41:586-598. [PMID: 28328100 DOI: 10.1002/cbin.10764] [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: 01/18/2017] [Accepted: 03/19/2017] [Indexed: 11/05/2022]
Abstract
At the early stage of atherosclerosis, neointima is formed due to the migration of vascular smooth muscle cells (VSMCs) from the media to the intima. VSMCs are surrounded by highly adhesive 3D matrices. They take specific strategies to cross various 3D matrices in the media, including heterogeneous collagen and mechanically strong basement membrane. Migration of VSMCs is potentially caused by biomechanical mechanism. Most in vitro studies focus on cell migration on 2D substrates in response to biochemical factors. How the cells move through 3D matrices under the action of mechanosensing machineries remains unexplored. In this review, we propose that several interesting tension-dependent machineries act as "tractor"-posterior myosin II accumulation, and "wrecker"-anterior podosome maintaining, to power VSMCs ahead. VSMCs embedded in 3D matrices may accumulate a minor myosin II isoform, myosin IIB, at the cell rear. Anisotropic myosin IIB distribution creates cell rear, polarizes cell body, pushes the nucleus and reshapes the cell body, and cooperates with a uniformly distributed myosin IIA to propel the cell forward. On the other hand, matrix digestion by podosome further promote the migration when the matrix becomes denser. Actomyosin tension activates Src to induce podosome in soft 3D matrices and retain the podosome integrity to steadily digest the matrix.
Collapse
Affiliation(s)
- Qingjia Chi
- Department of Mechanics and Engineering Structure, Wuhan University of Technology, Wuhan, Hubei, China
| | - Jieling Shan
- Department of Mechanics and Engineering Structure, Wuhan University of Technology, Wuhan, Hubei, China
| | - Xiaorong Ding
- Department of Electronic Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| | - Yazhou Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| | - Dongyu Jia
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), State and Local Joint Engineering Laboratory for Vascular Implants (Chongqing), Bioengineering College of Chongqing University, Chongqing, China
| |
Collapse
|
67
|
Coelho NM, Arora PD, van Putten S, Boo S, Petrovic P, Lin AX, Hinz B, McCulloch CA. Discoidin Domain Receptor 1 Mediates Myosin-Dependent Collagen Contraction. Cell Rep 2017; 18:1774-1790. [DOI: 10.1016/j.celrep.2017.01.061] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 12/20/2016] [Accepted: 01/24/2017] [Indexed: 01/04/2023] Open
|
68
|
Cyron CJ, Humphrey JD. Growth and Remodeling of Load-Bearing Biological Soft Tissues. MECCANICA 2017; 52:645-664. [PMID: 28286348 PMCID: PMC5342900 DOI: 10.1007/s11012-016-0472-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The past two decades reveal a growing role of continuum biomechanics in understanding homeostasis, adaptation, and disease progression in soft tissues. In this paper, we briefly review the two primary theoretical approaches for describing mechano-regulated soft tissue growth and remodeling on the continuum level as well as hybrid approaches that attempt to combine the advantages of these two approaches while avoiding their disadvantages. We also discuss emerging concepts, including that of mechanobiological stability. Moreover, to motivate and put into context the different theoretical approaches, we briefly review findings from mechanobiology that show the importance of mass turnover and the prestressing of both extant and new extracellular matrix in most cases of growth and remodeling. For illustrative purposes, these concepts and findings are discussed, in large part, within the context of two load-bearing, collagen dominated soft tissues - tendons/ligaments and blood vessels. We conclude by emphasizing further examples, needs, and opportunities in this exciting field of modeling soft tissues.
Collapse
Affiliation(s)
- C J Cyron
- Institute for Computational Mechanics, Technische Universität München, Garching, Germany
| | - J D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
69
|
Chang SJ, Chen YC, Yang CH, Huang SC, Huang HK, Li CC, Harn HIC, Chiu WT. Revealing the three dimensional architecture of focal adhesion components to explain Ca 2+-mediated turnover of focal adhesions. Biochim Biophys Acta Gen Subj 2017; 1861:624-635. [PMID: 28063985 DOI: 10.1016/j.bbagen.2017.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 12/07/2016] [Accepted: 01/03/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Focal adhesions (FAs) are large, dynamic protein complexes located close to the plasma membrane, which serve as the mechanical linkages and a biochemical signaling hub of cells. The coordinated and dynamic regulation of focal adhesion is required for cell migration. Degradation, or turnover, of FAs is a major event at the trailing edge of a migratory cell, and is mediated by Ca2+/calpain-dependent proteolysis and disassembly. Here, we investigated how Ca2+ influx induces cascades of FA turnover in living cells. METHODS Images obtained with a total internal reflection fluorescence microscope (TIRFM) showed that Ca2+ ions induce different processes in the FA molecules focal adhesion kinase (FAK), paxillin, vinculin, and talin. Three mutated calpain-resistant FA molecules, FAK-V744G, paxillin-S95G, and talin-L432G, were used to clarify the role of each FA molecule in FA turnover. RESULTS Vinculin was resistant to degradation and was not significantly affected by the presence of mutated calpain-resistant FA molecules. In contrast, talin was more sensitive to calpain-mediated turnover than the other molecules. Three-dimensional (3D) fluorescence imaging and immunoblotting demonstrated that outer FA molecules were more sensitive to calpain-mediated proteolysis than internal FA molecules. Furthermore, cell contraction is not involved in degradation of FA. CONCLUSIONS These results suggest that Ca2+-mediated degradation of FAs was mediated by both proteolysis and disassembly. The 3D architecture of FAs is related to the different dynamics of FA molecule degradation during Ca2+-mediated FA turnover. GENERAL SIGNIFICANCE This study will help us to clearly understand the underlying mechanism of focal adhesion turnover by Ca2+.
Collapse
Affiliation(s)
- Shu-Jing Chang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan
| | - Ying-Chi Chen
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan
| | - Chi-Hsun Yang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan
| | - Soon-Cen Huang
- Department of Obstetrics and Gynecology, Chi Mei Medical Center, Liouying Campus, Tainan 736, Taiwan
| | - Ho-Kai Huang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan
| | - Chun-Chun Li
- Department of Life Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Hans I-Chen Harn
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701, Taiwan; Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 701, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan 701, Taiwan.
| |
Collapse
|
70
|
Hirata H, Ku WC, Yip AK, Ursekar CP, Kawauchi K, Roy A, Guo AK, Vedula SRK, Harada I, Chiam KH, Ishihama Y, Lim CT, Sawada Y, Sokabe M. MEKK1-dependent phosphorylation of calponin-3 tunes cell contractility. J Cell Sci 2016; 129:3574-3582. [PMID: 27528401 DOI: 10.1242/jcs.189415] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 08/10/2016] [Indexed: 12/30/2022] Open
Abstract
MEKK1 (also known as MAP3K1), which plays a major role in MAPK signaling, has been implicated in mechanical processes in cells, such as migration. Here, we identify the actin-binding protein calponin-3 as a new MEKK1 substrate in the signaling that regulates actomyosin-based cellular contractility. MEKK1 colocalizes with calponin-3 at the actin cytoskeleton and phosphorylates it, leading to an increase in the cell-generated traction stress. MEKK1-mediated calponin-3 phosphorylation is attenuated by the inhibition of myosin II activity, the disruption of actin cytoskeletal integrity and adhesion to soft extracellular substrates, whereas it is enhanced upon cell stretching. Our results reveal the importance of the MEKK1-calponin-3 signaling pathway to cell contractility.
Collapse
Affiliation(s)
- Hiroaki Hirata
- Mechanobiology Institute, National University of Singapore, 117411 Singapore
| | - Wei-Chi Ku
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Ai Kia Yip
- A*STAR Bioinformatics Institute, 138671 Singapore
| | | | - Keiko Kawauchi
- Mechanobiology Institute, National University of Singapore, 117411 Singapore
| | - Amrita Roy
- Mechanobiology Institute, National University of Singapore, 117411 Singapore
| | - Alvin Kunyao Guo
- Mechanobiology Institute, National University of Singapore, 117411 Singapore
| | | | - Ichiro Harada
- Locomotive Syndrome Research Institute, Nadogaya Hospital, Kashiwa 277-0032, Japan Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Keng-Hwee Chiam
- Mechanobiology Institute, National University of Singapore, 117411 Singapore A*STAR Bioinformatics Institute, 138671 Singapore
| | - Yasushi Ishihama
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, 117411 Singapore Department of Biomedical Engineering, National University of Singapore, 117583 Singapore
| | - Yasuhiro Sawada
- Mechanobiology Institute, National University of Singapore, 117411 Singapore Locomotive Syndrome Research Institute, Nadogaya Hospital, Kashiwa 277-0032, Japan Department of Biological Sciences, National University of Singapore, 117543 Singapore
| | - Masahiro Sokabe
- Mechanobiology Institute, National University of Singapore, 117411 Singapore Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
71
|
Hwang MP, Subbiah R, Kim IG, Lee KE, Park J, Kim SH, Park K. Approximating bone ECM: Crosslinking directs individual and coupled osteoblast/osteoclast behavior. Biomaterials 2016; 103:22-32. [PMID: 27376556 DOI: 10.1016/j.biomaterials.2016.06.052] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/20/2016] [Accepted: 06/20/2016] [Indexed: 11/30/2022]
Abstract
Osteoblast and osteoclast communication (i.e. osteocoupling) is an intricate process, in which the biophysical profile of bone ECM is an aggregate product of their activities. While the effect of microenvironmental cues on osteoblast and osteoclast maturation has been resolved into individual variables (e.g. stiffness or topography), a single cue can be limited with regards to reflecting the full biophysical scope of natural bone ECM. Additionally, the natural modulation of bone ECM, which involves collagenous fibril and elastin crosslinking via lysyl oxidase, has yet to be reflected in current synthetic platforms. Here, we move beyond traditional substrates and use cell-derived ECM to examine individual and coupled osteoblast and osteoclast behavior on a physiological platform. Specifically, preosteoblast-derived ECM is crosslinked with genipin, a biocompatible crosslinker, to emulate physiological lysyl oxidase-mediated ECM crosslinking. We demonstrate that different concentrations of genipin yield changes to ECM density, stiffness, and roughness while retaining biocompatibility. By approximating various bone ECM profiles, we examine how individual and coupled osteoblast and osteoclast behavior are affected. Ultimately, we demonstrate an increase in osteoblast and osteoclast differentiation on compact and loose ECM, respectively, and identify ECM crosslinking density as an underlying force in osteocoupling behavior.
Collapse
Affiliation(s)
- Mintai P Hwang
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ramesh Subbiah
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Department of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - In Gul Kim
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Kyung Eun Lee
- Advanced Analysis Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Jimin Park
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Sang Heon Kim
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Department of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Department of Biomedical Engineering, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea.
| |
Collapse
|
72
|
Wei XH, Lin SS, Liu Y, Zhao RP, Khan GJ, Du HZ, Mao TT, Yu BY, Li RM, Yuan ST, Sun L. DT-13 attenuates human lung cancer metastasis via regulating NMIIA activity under hypoxia condition. Oncol Rep 2016; 36:991-9. [DOI: 10.3892/or.2016.4879] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 03/08/2016] [Indexed: 11/05/2022] Open
|
73
|
Chen YF, Hsu KF, Shen MR. The store-operated Ca 2+ entry-mediated signaling is important for cancer spread. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1427-35. [DOI: 10.1016/j.bbamcr.2015.11.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 11/17/2015] [Accepted: 11/25/2015] [Indexed: 12/31/2022]
|
74
|
Measurements of Myosin-II Motor Activity During Cytokinesis in Fission Yeast. Methods Mol Biol 2016; 1369:137-50. [PMID: 26519311 DOI: 10.1007/978-1-4939-3145-3_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Fission yeast myosin-II (Myo2p) represents the critical actin-based motor protein that drives actomyosin ring assembly and constriction during cytokinesis. We detail three different methods to measure Myo2p motor function. Actin-activated ATPases provide a readout of actomyosin ATPase motor activity in a bulk assay; actin filament motility assays reveal the speed and efficiency of myosin-driven actin filament gliding (when motors are anchored); myosin-bead motility assays reveal the speed and efficiency of myosin ensembles traveling along actin filaments (when actin is anchored). Collectively, these methods allow us to combine the standard in vivo approaches common to fission yeast with in vitro biochemical methods to learn more about the mechanistic action of myosin-II during cytokinesis.
Collapse
|
75
|
Canver AC, Ngo O, Urbano RL, Clyne AM. Endothelial directed collective migration depends on substrate stiffness via localized myosin contractility and cell-matrix interactions. J Biomech 2016; 49:1369-1380. [DOI: 10.1016/j.jbiomech.2015.12.037] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 11/25/2015] [Accepted: 12/16/2015] [Indexed: 01/05/2023]
|
76
|
Lourenço T, Paes de Faria J, Bippes CA, Maia J, Lopes-da-Silva JA, Relvas JB, Grãos M. Modulation of oligodendrocyte differentiation and maturation by combined biochemical and mechanical cues. Sci Rep 2016; 6:21563. [PMID: 26879561 PMCID: PMC4754901 DOI: 10.1038/srep21563] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 01/25/2016] [Indexed: 01/17/2023] Open
Abstract
Extracellular matrix (ECM) proteins play a key role during oligodendrogenesis. While fibronectin (FN) is involved in the maintenance and proliferation of oligodendrocyte progenitor cells (OPCs), merosin (MN) promotes differentiation into oligodendrocytes (OLs). Mechanical properties of the ECM also seem to affect OL differentiation, hence this study aimed to clarify the impact of combined biophysical and biochemical elements during oligodendrocyte differentiation and maturation using synthetic elastic polymeric ECM-like substrates. CG-4 cells presented OPC- or OL-like morphology in response to brain-compliant substrates functionalised with FN or MN, respectively. The expression of the differentiation and maturation markers myelin basic protein — MBP — and proteolipid protein — PLP — (respectively) by primary rat oligodendrocytes was enhanced in presence of MN, but only on brain-compliant conditions, considering the distribution (MBP) or amount (PLP) of the protein. It was also observed that maturation of OLs was attained earlier (by assessing PLP expression) by cells differentiated on MN-functionalised brain-compliant substrates than on standard culture conditions. Moreover, the combination of MN and substrate compliance enhanced the maturation and morphological complexity of OLs. Considering the distinct degrees of stiffness tested ranging within those of the central nervous system, our results indicate that 6.5 kPa is the most suitable rigidity for oligodendrocyte differentiation.
Collapse
Affiliation(s)
- Tânia Lourenço
- Biocant, Technology Transfer Association, Cantanhede, Portugal.,Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Joana Paes de Faria
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | | | - João Maia
- Chemical Engineering Department, Faculty of Science and Technology, University of Coimbra, Coimbra, Portugal
| | | | - João B Relvas
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - Mário Grãos
- Biocant, Technology Transfer Association, Cantanhede, Portugal.,Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
77
|
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
|
78
|
Jerrell RJ, Parekh A. Matrix rigidity differentially regulates invadopodia activity through ROCK1 and ROCK2. Biomaterials 2016; 84:119-129. [PMID: 26826790 DOI: 10.1016/j.biomaterials.2016.01.028] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 01/12/2016] [Accepted: 01/14/2016] [Indexed: 01/17/2023]
Abstract
ROCK activity increases due to ECM rigidity in the tumor microenvironment and promotes a malignant phenotype via actomyosin contractility. Invasive migration is facilitated by actin-rich adhesive protrusions known as invadopodia that degrade the ECM. Invadopodia activity is dependent on matrix rigidity and contractile forces suggesting that mechanical factors may regulate these subcellular structures through ROCK-dependent actomyosin contractility. However, emerging evidence indicates that the ROCK1 and ROCK2 isoforms perform different functions in cells suggesting that alternative mechanisms may potentially regulate rigidity-dependent invadopodia activity. In this study, we found that matrix rigidity drives ROCK signaling in cancer cells but that ROCK1 and ROCK2 differentially regulate invadopodia activity through separate signaling pathways via contractile (NM II) and non-contractile (LIMK) mechanisms. These data suggest that the mechanical rigidity of the tumor microenvironment may drive ROCK signaling through distinct pathways to enhance the invasive migration required for cancer progression and metastasis.
Collapse
Affiliation(s)
- Rachel J Jerrell
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Aron Parekh
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
79
|
Fujii Y, Funakoshi T, Unuma K, Noritake K, Aki T, Uemura K. Hydrogen sulfide donor NaHS induces death of alveolar epithelial L2 cells that is associated with cellular shrinkage, transgelin expression and myosin phosphorylation. J Toxicol Sci 2016; 41:645-54. [DOI: 10.2131/jts.41.645] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Yusuke Fujii
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Takeshi Funakoshi
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Kana Unuma
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Kanako Noritake
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Toshihiko Aki
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Koichi Uemura
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| |
Collapse
|
80
|
Kim K, Ossipova O, Sokol SY. Neural crest specification by inhibition of the ROCK/Myosin II pathway. Stem Cells 2015; 33:674-85. [PMID: 25346532 DOI: 10.1002/stem.1877] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 08/13/2014] [Accepted: 09/13/2014] [Indexed: 01/14/2023]
Abstract
Neural crest is a population of multipotent progenitor cells that form at the border of neural and non-neural ectoderm in vertebrate embryos, and undergo epithelial-mesenchymal transition and migration. According to the traditional view, the neural crest is specified in early embryos by signaling molecules including BMP, FGF, and Wnt proteins. Here, we identify a novel signaling pathway leading to neural crest specification, which involves Rho-associated kinase (ROCK) and its downstream target nonmuscle Myosin II. We show that ROCK inhibitors promote differentiation of human embryonic stem cells (hESCs) into neural crest-like progenitors (NCPs) that are characterized by specific molecular markers and ability to differentiate into multiple cell types, including neurons, chondrocytes, osteocytes, and smooth muscle cells. Moreover, inhibition of Myosin II was sufficient for generating NCPs at high efficiency. Whereas Myosin II has been previously implicated in the self-renewal and survival of hESCs, we demonstrate its role in neural crest development during ESC differentiation. Inhibition of this pathway in Xenopus embryos expanded neural crest in vivo, further indicating that neural crest specification is controlled by ROCK-dependent Myosin II activity. We propose that changes in cell morphology in response to ROCK and Myosin II inhibition initiate mechanical signaling leading to neural crest fates.
Collapse
Affiliation(s)
- Kyeongmi Kim
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | |
Collapse
|
81
|
Parekh A, Weaver AM. Regulation of invadopodia by mechanical signaling. Exp Cell Res 2015; 343:89-95. [PMID: 26546985 DOI: 10.1016/j.yexcr.2015.10.038] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 10/31/2015] [Indexed: 12/15/2022]
Abstract
Mechanical rigidity in the tumor microenvironment is associated with a high risk of tumor formation and aggressiveness. Adhesion-based signaling driven by a rigid microenvironment is thought to facilitate invasion and migration of cancer cells away from primary tumors. Proteolytic degradation of extracellular matrix (ECM) is a key component of this process and is mediated by subcellular actin-rich structures known as invadopodia. Both ECM rigidity and cellular traction stresses promote invadopodia formation and activity, suggesting a role for these structures in mechanosensing. The presence and activity of mechanosensitive adhesive and signaling components at invadopodia further indicates the potential for these structures to utilize myosin-dependent forces to probe and remodel their ECM environments. Here, we provide a brief review of the role of adhesion-based mechanical signaling in controlling invadopodia and invasive cancer behavior.
Collapse
Affiliation(s)
- Aron Parekh
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN 37232 USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232 USA.
| | - Alissa M Weaver
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232 USA; Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232 USA; Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232 USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232 USA.
| |
Collapse
|
82
|
Park YM. Oxidized LDL induces phosphorylation of non-muscle myosin IIA heavy chain in macrophages. BMB Rep 2015; 48:48-53. [PMID: 25322953 PMCID: PMC4345642 DOI: 10.5483/bmbrep.2015.48.1.186] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 09/23/2014] [Accepted: 10/16/2014] [Indexed: 11/20/2022] Open
Abstract
Oxidized LDL (oxLDL) performs critical roles in atherosclerosis by inducing macrophage foam cell formation and promoting inflammation. There have been reports showing that oxLDL modulates macrophage cytoskeletal functions for oxLDL uptake and trapping, however, the precise mechanism has not been clearly elucidated. Our study examined the effect of oxLDL on non-muscle myosin heavy chain IIA (MHC-IIA) in macrophages. We demonstrated that oxLDL induces phosphorylation of MHC-IIA (Ser1917) in peritoneal macrophages from wild-type mice and THP-1, a human monocytic cell line, but not in macrophages deficient for CD36, a scavenger receptor for oxLDL. Protein kinase C (PKC) inhibitor-treated macrophages did not undergo the oxLDL-induced MHC-IIA phosphorylation. Our immunoprecipitation revealed that oxLDL increased physical association between PKC and MHC-IIA, supporting the role of PKC in this process. We conclude that oxLDL via CD36 induces PKC-mediated MHC-IIA (Ser1917) phosphorylation and this may affect oxLDL-induced functions of macrophages involved in atherosclerosis.
Collapse
Affiliation(s)
- Young Mi Park
- Department of Molecular Medicine, Ewha Womans University School of Medicine; Global Top 5 Research Program, Ewha Womans University, Seoul 158-710, Korea
| |
Collapse
|
83
|
Shadrin IY, Yoon W, Li L, Shepherd N, Bursac N. Rapid fusion between mesenchymal stem cells and cardiomyocytes yields electrically active, non-contractile hybrid cells. Sci Rep 2015; 5:12043. [PMID: 26159124 PMCID: PMC4498233 DOI: 10.1038/srep12043] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/12/2015] [Indexed: 12/26/2022] Open
Abstract
Cardiac cell therapies involving bone marrow-derived human mesenchymal stem cells (hMSCs) have shown promising results, although their mechanisms of action are still poorly understood. Here, we investigated direct interactions between hMSCs and cardiomyocytes in vitro. Using a genetic Ca2+ indicator gCaMP3 to efficiently label hMSCs in co-cultures with neonatal rat ventricular myocytes (NRVMs), we determined that 25–40% of hMSCs (from 4 independent donors) acquired periodic Ca2+ transients and cardiac markers through spontaneous fusion with NRVMs. Sharp electrode and voltage-clamp recordings in fused cells showed action potential properties and Ca2+ current amplitudes in between those of non-fused hMSCs and NRVMs. Time-lapse video-microscopy revealed the first direct evidence of active fusion between hMSCs and NRVMs within several hours of co-culture. Application of blebbistatin, nifedipine or verapamil caused complete and reversible inhibition of fusion, suggesting potential roles for actomyosin bridging and Ca2+ channels in the fusion process. Immunostaining for Cx43, Ki67, and sarcomeric α-actinin showed that fused cells remain strongly coupled to surrounding NRVMs, but downregulate sarcomeric structures over time, acquiring a non-proliferative and non-contractile phenotype. Overall, these results describe the phenotype and mechanisms of hybrid cell formation via fusion of hMSCs and cardiomyocytes with potential implications for cardiac cell therapy.
Collapse
Affiliation(s)
- Ilya Y Shadrin
- Department of Biomedical Engineering, Duke University, Durham, NC
| | - Woohyun Yoon
- Department of Biomedical Engineering, Duke University, Durham, NC
| | - Liqing Li
- Department of Biomedical Engineering, Duke University, Durham, NC
| | - Neal Shepherd
- Department of Biomedical Engineering, Duke University, Durham, NC
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC
| |
Collapse
|
84
|
Abstract
Neuronal growth cones are exquisite sensory-motor machines capable of transducing features contacted in their local extracellular environment into guided process extension during development. Extensive research has shown that chemical ligands activate cell surface receptors on growth cones leading to intracellular signals that direct cytoskeletal changes. However, the environment also provides mechanical support for growth cone adhesion and traction forces that stabilize leading edge protrusions. Interestingly, recent work suggests that both the mechanical properties of the environment and mechanical forces generated within growth cones influence axon guidance. In this review we discuss novel molecular mechanisms involved in growth cone force production and detection, and speculate how these processes may be necessary for the development of proper neuronal morphogenesis.
Collapse
Affiliation(s)
- Patrick C Kerstein
- Neuroscience Training Program, Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison Madison, WI, USA
| | - Robert H Nichol
- Neuroscience Training Program, Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison Madison, WI, USA
| | - Timothy M Gomez
- Neuroscience Training Program, Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison Madison, WI, USA
| |
Collapse
|
85
|
Hindman B, Goeckeler Z, Sierros K, Wysolmerski R. Non-Muscle Myosin II Isoforms Have Different Functions in Matrix Rearrangement by MDA-MB-231 Cells. PLoS One 2015; 10:e0131920. [PMID: 26136073 PMCID: PMC4489869 DOI: 10.1371/journal.pone.0131920] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/08/2015] [Indexed: 01/15/2023] Open
Abstract
The role of a stiffening extra-cellular matrix (ECM) in cancer progression is documented but poorly understood. Here we use a conditioning protocol to test the role of nonmuscle myosin II isoforms in cell mediated ECM arrangement using collagen constructs seeded with breast cancer cells expressing shRNA targeted to either the IIA or IIB heavy chain isoform. While there are several methods available to measure changes in the biophysical characteristics of the ECM, we wanted to use a method which allows for the measurement of global stiffness changes as well as a dynamic response from the sample over time. The conditioning protocol used allows the direct measurement of ECM stiffness. Using various treatments, it is possible to determine the contribution of various construct and cellular components to the overall construct stiffness. Using this assay, we show that both the IIA and IIB isoforms are necessary for efficient matrix remodeling by MDA-MB-231 breast cancer cells, as loss of either isoform changes the stiffness of the collagen constructs as measured using our conditioning protocol. Constructs containing only collagen had an elastic modulus of 0.40 Pascals (Pa), parental MDA-MB-231 constructs had an elastic modulus of 9.22 Pa, while IIA and IIB KD constructs had moduli of 3.42 and 7.20 Pa, respectively. We also calculated the cell and matrix contributions to the overall sample elastic modulus. Loss of either myosin isoform resulted in decreased cell stiffness, as well as a decrease in the stiffness of the cell-altered collagen matrices. While the total construct modulus for the IIB KD cells was lower than that of the parental cells, the IIB KD cell-altered matrices actually had a higher elastic modulus than the parental cell-altered matrices (4.73 versus 4.38 Pa). These results indicate that the IIA and IIB heavy chains play distinct and non-redundant roles in matrix remodeling.
Collapse
Affiliation(s)
- Bridget Hindman
- Mary Babb Randolph Cancer Center, West Virginia University, Robert C. Byrd Health Sciences Center, Morgantown, West Virginia, United States of America
| | - Zoe Goeckeler
- Center for Cardiovascular and Respiratory Diseases, West Virginia University, Robert C. Byrd Health Sciences Center, Morgantown, West Virginia, United States of America
| | - Kostas Sierros
- Mechanical and Aerospace Engineering, Statler College of Engineering and Mineral Resources, West Virginia University, Morgantown, West Virginia, United States of America
| | - Robert Wysolmerski
- Mary Babb Randolph Cancer Center, West Virginia University, Robert C. Byrd Health Sciences Center, Morgantown, West Virginia, United States of America
- Center for Cardiovascular and Respiratory Diseases, West Virginia University, Robert C. Byrd Health Sciences Center, Morgantown, West Virginia, United States of America
- * E-mail:
| |
Collapse
|
86
|
Zheng C, Wang J, Hao JH. Role of store-operated calcium entry in digestive system tumors. Shijie Huaren Xiaohua Zazhi 2015; 23:1609-1614. [DOI: 10.11569/wcjd.v23.i10.1609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Store-operated calcium entry (SOCE) is one of the important channels mediating extracellular Ca2+ entry. Stromal interaction molecule 1 (STIM1) protein in the membrane of endoplasmic reticulum and calcium release-activated calcium channel protein 1 (CRCM1/Orai1) protein in the plasma membrane are two key components of SOCE. Current research has gradually elucidated the mechanism of action of SOCE in tumors, and it has been demonstrated that SOCE plays an important role in digestive system tumors. This paper gives an overview of SOCE and reviews the current progress in understanding the role of SOCE in digestive system tumors.
Collapse
|
87
|
Sebé-Pedrós A, Grau-Bové X, Richards TA, Ruiz-Trillo I. Evolution and classification of myosins, a paneukaryotic whole-genome approach. Genome Biol Evol 2015; 6:290-305. [PMID: 24443438 PMCID: PMC3942036 DOI: 10.1093/gbe/evu013] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Myosins are key components of the eukaryotic cytoskeleton, providing motility for a broad diversity of cargoes. Therefore, understanding the origin and evolutionary history of myosin classes is crucial to address the evolution of eukaryote cell biology. Here, we revise the classification of myosins using an updated taxon sampling that includes newly or recently sequenced genomes and transcriptomes from key taxa. We performed a survey of eukaryotic genomes and phylogenetic analyses of the myosin gene family, reconstructing the myosin toolkit at different key nodes in the eukaryotic tree of life. We also identified the phylogenetic distribution of myosin diversity in terms of number of genes, associated protein domains and number of classes in each taxa. Our analyses show that new classes (i.e., paralogs) and domain architectures were continuously generated throughout eukaryote evolution, with a significant expansion of myosin abundance and domain architectural diversity at the stem of Holozoa, predating the origin of animal multicellularity. Indeed, single-celled holozoans have the most complex myosin complement among eukaryotes, with paralogs of most myosins previously considered animal specific. We recover a dynamic evolutionary history, with several lineage-specific expansions (e.g., the myosin III-like gene family diversification in choanoflagellates), convergence in protein domain architectures (e.g., fungal and animal chitin synthase myosins), and important secondary losses. Overall, our evolutionary scheme demonstrates that the ancestral eukaryote likely had a complex myosin repertoire that included six genes with different protein domain architectures. Finally, we provide an integrative and robust classification, useful for future genomic and functional studies on this crucial eukaryotic gene family.
Collapse
Affiliation(s)
- Arnau Sebé-Pedrós
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Passeig Marítim de la Barceloneta, Barcelona, Catalonia, Spain
| | | | | | | |
Collapse
|
88
|
Pasapera AM, Plotnikov SV, Fischer RS, Case LB, Egelhoff TT, Waterman CM. Rac1-dependent phosphorylation and focal adhesion recruitment of myosin IIA regulates migration and mechanosensing. Curr Biol 2014; 25:175-186. [PMID: 25544611 DOI: 10.1016/j.cub.2014.11.043] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 07/14/2014] [Accepted: 11/14/2014] [Indexed: 01/19/2023]
Abstract
BACKGROUND Cell migration requires coordinated formation of focal adhesions (FAs) and assembly and contraction of the actin cytoskeleton. Nonmuscle myosin II (MII) is a critical mediator of contractility and FA dynamics in cell migration. Signaling downstream of the small GTPase Rac1 also regulates FA and actin dynamics, but its role in regulation of MII during migration is less clear. RESULTS We found that Rac1 promotes association of MIIA with FA. Live-cell imaging showed that, whereas most MIIA at the leading edge assembled into dorsal contractile arcs, a substantial subset assembled in or was captured within maturing FA, and this behavior was promoted by active Rac1. Protein kinase C (PKC) activation was necessary and sufficient for integrin- and Rac1-dependent phosphorylation of MIIA heavy chain (HC) on serine1916 (S1916) and recruitment to FA. S1916 phosphorylation of MIIA HC and localization in FA was enhanced during cell spreading and ECM stiffness mechanosensing, suggesting upregulation of this pathway during physiological Rac1 activation. Phosphomimic and nonphosphorylatable MIIA HC mutants demonstrated that S1916 phosphorylation was necessary and sufficient for the capture and assembly of MIIA minifilaments in FA. S1916 phosphorylation was also sufficient to promote the rapid assembly of FAs to enhance cell migration and for the modulation of traction force, spreading, and migration by ECM stiffness. CONCLUSIONS Our study reveals for the first time that Rac1 and integrin activation regulates MIIA HC phosphorylation through a PKC-dependent mechanism that promotes MIIA association with FAs and acts as a critical modulator of cell migration and mechanosensing.
Collapse
Affiliation(s)
- Ana M Pasapera
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sergey V Plotnikov
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Robert S Fischer
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lindsay B Case
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thomas T Egelhoff
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Clare M Waterman
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
89
|
DeMali KA, Sun X, Bui GA. Force transmission at cell-cell and cell-matrix adhesions. Biochemistry 2014; 53:7706-17. [PMID: 25474123 DOI: 10.1021/bi501181p] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
All cells are subjected to mechanical forces throughout their lifetimes. These forces are sensed by cell surface adhesion receptors and trigger robust actin cytoskeletal rearrangements and growth of the associated adhesion complex to counter the applied force. In this review, we discuss how integrins and cadherins sense force and transmit these forces into the cell interior. We focus on the complement of proteins each adhesion complex recruits to bear the force and the signal transduction pathways activated to allow the cell to tune its contractility. A discussion of the similarities, differences, and crosstalk between cadherin- and integrin-mediated force transmission is also presented.
Collapse
Affiliation(s)
- Kris A DeMali
- Department of Biochemistry and Interdisciplinary Program in Molecular and Cellular Biology, Roy J. and Lucille A. Carver College of Medicine , Iowa City, Iowa 52242, United States
| | | | | |
Collapse
|
90
|
You E, Park S, Kim D, Jung J, Ko P, Park CM, Rhee S. Role of the intracellular juxtamembrane domain of discoidin domain receptor 2 in focal adhesion formation. Anim Cells Syst (Seoul) 2014. [DOI: 10.1080/19768354.2014.969770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
91
|
Platelet mechanosensing of substrate stiffness during clot formation mediates adhesion, spreading, and activation. Proc Natl Acad Sci U S A 2014; 111:14430-5. [PMID: 25246564 DOI: 10.1073/pnas.1322917111] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
As platelets aggregate and activate at the site of vascular injury to stem bleeding, they are subjected to a myriad of biochemical and biophysical signals and cues. As clot formation ensues, platelets interact with polymerizing fibrin scaffolds, exposing platelets to a large range of mechanical microenvironments. Here, we show for the first time (to our knowledge) that platelets, which are anucleate cellular fragments, sense microenvironmental mechanical properties, such as substrate stiffness, and transduce those cues into differential biological signals. Specifically, as platelets mechanosense the stiffness of the underlying fibrin/fibrinogen substrate, increasing substrate stiffness leads to increased platelet adhesion and spreading. Importantly, adhesion on stiffer substrates also leads to higher levels of platelet activation, as measured by integrin αIIbβ3 activation, α-granule secretion, and procoagulant activity. Mechanistically, we determined that Rac1 and actomyosin activity mediate substrate stiffness-dependent platelet adhesion, spreading, and activation to different degrees. This capability of platelets to mechanosense microenvironmental cues in a growing thrombus or hemostatic plug and then mechanotransduce those cues into differential levels of platelet adhesion, spreading, and activation provides biophysical insight into the underlying mechanisms of platelet aggregation and platelet activation heterogeneity during thrombus formation.
Collapse
|
92
|
Gammon DB, Duraffour S, Rozelle DK, Hehnly H, Sharma R, Sparks ME, West CC, Chen Y, Moresco JJ, Andrei G, Connor JH, Conte D, Gundersen-Rindal DE, Marshall WL, Yates JR, Silverman N, Mello CC. A single vertebrate DNA virus protein disarms invertebrate immunity to RNA virus infection. eLife 2014; 3:e02910. [PMID: 24966209 PMCID: PMC4112549 DOI: 10.7554/elife.02910] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 06/25/2014] [Indexed: 12/12/2022] Open
Abstract
Virus-host interactions drive a remarkable diversity of immune responses and countermeasures. We found that two RNA viruses with broad host ranges, vesicular stomatitis virus (VSV) and Sindbis virus (SINV), are completely restricted in their replication after entry into Lepidopteran cells. This restriction is overcome when cells are co-infected with vaccinia virus (VACV), a vertebrate DNA virus. Using RNAi screening, we show that Lepidopteran RNAi, Nuclear Factor-κB, and ubiquitin-proteasome pathways restrict RNA virus infection. Surprisingly, a highly conserved, uncharacterized VACV protein, A51R, can partially overcome this virus restriction. We show that A51R is also critical for VACV replication in vertebrate cells and for pathogenesis in mice. Interestingly, A51R colocalizes with, and stabilizes, host microtubules and also associates with ubiquitin. We show that A51R promotes viral protein stability, possibly by preventing ubiquitin-dependent targeting of viral proteins for destruction. Importantly, our studies reveal exciting new opportunities to study virus-host interactions in experimentally-tractable Lepidopteran systems.
Collapse
Affiliation(s)
- Don B Gammon
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, United States
| | | | - Daniel K Rozelle
- Department of Microbiology, Boston University, Boston, United States
| | - Heidi Hehnly
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Rita Sharma
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, United States
- Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, United States
| | - Michael E Sparks
- Agricultural Research Service, United States Department of Agriculture, Beltsville, United States
| | - Cara C West
- Department of Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Ying Chen
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, United States
| | - James J Moresco
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, United States
| | - Graciela Andrei
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - John H Connor
- Department of Microbiology, Boston University, Boston, United States
| | - Darryl Conte
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, United States
| | - Dawn E Gundersen-Rindal
- Agricultural Research Service, United States Department of Agriculture, Beltsville, United States
| | - William L Marshall
- Department of Medicine, University of Massachusetts Medical School, Worcester, United States
| | - John R Yates
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, United States
| | - Neal Silverman
- Department of Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Craig C Mello
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, United States
- Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, United States
| |
Collapse
|
93
|
Morin TR, Ghassem-Zadeh SA, Lee J. Traction force microscopy in rapidly moving cells reveals separate roles for ROCK and MLCK in the mechanics of retraction. Exp Cell Res 2014; 326:280-94. [PMID: 24786318 DOI: 10.1016/j.yexcr.2014.04.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 04/15/2014] [Accepted: 04/18/2014] [Indexed: 01/13/2023]
Abstract
Retraction is a major rate-limiting step in cell motility, particularly in slow moving cell types that form large stable adhesions. Myosin II dependent contractile forces are thought to facilitate detachment by physically pulling up the rear edge. However, retraction can occur in the absence of myosin II activity in cell types that form small labile adhesions. To investigate the role of contractile force generation in retraction, we performed traction force microscopy during the movement of fish epithelial keratocytes. By correlating changes in local traction stress at the rear with the area retracted, we identified four distinct modes of retraction. "Recoil" retractions are preceded by a rise in local traction stress, while rear edge is temporarily stuck, followed by a sharp drop in traction stress upon detachment. This retraction type was most common in cells generating high average traction stress. In "pull" type retractions local traction stress and area retracted increase concomitantly. This was the predominant type of retraction in keratocytes and was observed mostly in cells generating low average traction stress. "Continuous" type retractions occur without any detectable change in traction stress, and are seen in cells generating low average traction stress. In contrast, to many other cell types, "release" type retractions occur in keratocytes following a decrease in local traction stress. Our identification of distinct modes of retraction suggests that contractile forces may play different roles in detachment that are related to rear adhesion strength. To determine how the regulation of contractility via MLCK or Rho kinase contributes to the mechanics of detachment, inhibitors were used to block or augment these pathways. Modulation of MLCK activity led to the most rapid change in local traction stress suggesting its importance in regulating attachment strength. Surprisingly, Rho kinase was not required for detachment, but was essential for localizing retraction to the rear. We suggest that in keratocytes MLCK and Rho kinase play distinct, complementary roles in the respective temporal and spatial control of rear detachment that is essential for maintaining rapid motility.
Collapse
Affiliation(s)
- Timothy R Morin
- Department of Molecular and Cell Biology, University of Connecticut, 91 North Eagleville Road, Storrs, CT 06269, USA
| | - Sean A Ghassem-Zadeh
- Department of Molecular and Cell Biology, University of Connecticut, 91 North Eagleville Road, Storrs, CT 06269, USA
| | - Juliet Lee
- Department of Molecular and Cell Biology, University of Connecticut, 91 North Eagleville Road, Storrs, CT 06269, USA.
| |
Collapse
|
94
|
The different roles of myosin IIA and myosin IIB in contraction of 3D collagen matrices by human fibroblasts. Exp Cell Res 2014; 326:295-306. [PMID: 24768700 DOI: 10.1016/j.yexcr.2014.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/09/2014] [Accepted: 04/14/2014] [Indexed: 02/06/2023]
Abstract
Contraction of 3D collagen matrices by fibroblasts frequently is used as an in vitro model of wound closure. Different iterations of the model - all conventionally referred to as "contraction" - involve different morphological patterns. During floating matrix contraction, cells initially are round without stress fibers and subsequently undergo spreading. During stressed matrix contraction, cells initially are spread with stress fibers and subsequently undergo shortening. In the current studies, we used siRNA silencing of myosin IIA (MyoIIA) and myosin IIB (MyoIIB) to test the roles of myosin II isoforms in fibroblast interactions with 3D collagen matrices and collagen matrix contraction. We found that MyoIIA but not MyoIIB was required for cellular global inward contractile force, formation of actin stress fibers, and morphogenic cell clustering. Stressed matrix contraction required MyoIIA but not MyoIIB. Either MyoIIA or MyoIIB was sufficient for floating matrix contraction (FMC) stimulated by platelet-derived growth factor. Neither MyoIIA or MyoIIB was necessary for FMC stimulated by serum. Our findings suggest that myosin II-dependent motor mechanisms for collagen translocation during extracellular matrix remodeling differ depending on cell tension and growth factor stimulation.
Collapse
|
95
|
Gitik M, Kleinhaus R, Hadas S, Reichert F, Rotshenker S. Phagocytic receptors activate and immune inhibitory receptor SIRPα inhibits phagocytosis through paxillin and cofilin. Front Cell Neurosci 2014; 8:104. [PMID: 24795566 PMCID: PMC3997012 DOI: 10.3389/fncel.2014.00104] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 03/24/2014] [Indexed: 11/13/2022] Open
Abstract
The innate immune function of phagocytosis of apoptotic cells, tissue debris, pathogens, and cancer cells is essential for homeostasis, tissue repair, fighting infection, and combating malignancy. Phagocytosis is carried out in the central nervous system (CNS) by resident microglia and in both CNS and peripheral nervous system by recruited macrophages. While phagocytosis proceeds, bystander healthy cells protect themselves by sending a "do not eat me" message to phagocytes as CD47 on their surface ligates immune inhibitory receptor SIRPα on the surface of phagocytes and SIRPα then produces the signaling which inhibits phagocytosis. This helpful mechanism becomes harmful when tissue debris and unhealthy cells inhibit their own phagocytosis by employing the same mechanism. However, the inhibitory signaling that SIRPα produces has not been fully revealed. We focus here on how SIRPα inhibits the phagocytosis of the tissue debris "degenerated myelin" which hinders repair in axonal injury and neurodegenerative diseases. We tested whether SIRPα inhibits phagocytosis by regulating cytoskeleton function through paxillin and cofilin since (a) the cytoskeleton generates the mechanical forces that drive phagocytosis and (b) both paxillin and cofilin control cytoskeleton function. Paxillin and cofilin were transiently activated in microglia as phagocytosis was activated. In contrast, paxillin and cofilin were continuously activated and phagocytosis augmented in microglia in which SIRPα expression was knocked-down by SIRPα-shRNA. Further, levels of phagocytosis, paxillin activation, and cofilin activation positively correlated with one another. Taken together, these observations suggest a novel mechanism whereby paxillin and cofilin are targeted to control phagocytosis by both the activating signaling that phagocytic receptors produce by promoting the activation of paxillin and cofilin and the inhibiting signaling that immune inhibitory SIRPα produces by promoting the inactivation of paxillin and cofilin.
Collapse
Affiliation(s)
- Miri Gitik
- Department of Medical Neurobiology, Institute for Medical Research Israel–Canada, Faculty of Medicine, Hebrew UniversityJerusalem, Israel
| | - Rachel Kleinhaus
- Department of Medical Neurobiology, Institute for Medical Research Israel–Canada, Faculty of Medicine, Hebrew UniversityJerusalem, Israel
| | - Smadar Hadas
- Department of Medical Neurobiology, Institute for Medical Research Israel–Canada, Faculty of Medicine, Hebrew UniversityJerusalem, Israel
| | - Fanny Reichert
- Department of Medical Neurobiology, Institute for Medical Research Israel–Canada, Faculty of Medicine, Hebrew UniversityJerusalem, Israel
| | - Shlomo Rotshenker
- Department of Medical Neurobiology, Institute for Medical Research Israel–Canada, Faculty of Medicine, Hebrew UniversityJerusalem, Israel
- Brain Disease Research Center, Institute for Medical Research Israel–Canada, Faculty of Medicine, Hebrew UniversityJerusalem, Israel
| |
Collapse
|
96
|
Chi Q, Yin T, Gregersen H, Deng X, Fan Y, Zhao J, Liao D, Wang G. Rear actomyosin contractility-driven directional cell migration in three-dimensional matrices: a mechano-chemical coupling mechanism. J R Soc Interface 2014; 11:20131072. [PMID: 24647903 DOI: 10.1098/rsif.2013.1072] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Cell migration is of vital importance in many biological processes, including organismal development, immune response and development of vascular diseases. For instance, migration of vascular smooth muscle cells from the media to intima is an essential part of the development of atherosclerosis and restenosis after stent deployment. While it is well characterized that cells use actin polymerization at the leading edge to propel themselves to move on two-dimensional substrates, the migration modes of cells in three-dimensional matrices relevant to in vivo environments remain unclear. Intracellular tension, which is created by myosin II activity, fulfils a vital role in regulating cell migration. We note that there is compelling evidence from theoretical and experimental work that myosin II accumulates at the cell rear, either isoform-dependent or -independent, leading to three-dimensional migration modes driven by posterior myosin II tension. The scenario is not limited to amoeboid migration, and it is also seen in mesenchymal migration in which a two-dimensional-like migration mode based on front protrusions is often expected, suggesting that there may exist universal underlying mechanisms. In this review, we aim to shed some light on how anisotropic myosin II localization induces cell motility in three-dimensional environments from a biomechanical view. We demonstrate an interesting mechanism where an interplay between mechanical myosin II recruitment and biochemical myosin II activation triggers directional migration in three-dimensional matrices. In the case of amoeboid three-dimensional migration, myosin II first accumulates at the cell rear to induce a slight polarization displayed as a uropod-like structure under the action of a tension-dependent mechanism. Subsequent biochemical signalling pathways initiate actomyosin contractility, producing traction forces on the adhesion system or creating prominent motile forces through blebbing activity, to drive cells to move. In mesenchymal three-dimensional migration, cells can also take advantage of the elastic properties of three-dimensional matrices to move. A minor myosin isoform, myosin IIB, is retained by relatively stiff three-dimensional matrices at the posterior side, then activated by signalling cascades, facilitating prominent cell polarization by establishing front-back polarity and creating cell rear. Myosin IIB initiates cell polarization and coordinates with the major isoform myosin IIA-assembled stress fibres, to power the directional migration of cells in the three-dimensional matrix.
Collapse
Affiliation(s)
- Qingjia Chi
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education; Chongqing Engineering Laboratory in Vascular Implants; Bioengineering College of Chongqing University, , Chongqing, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
97
|
Schulze N, Graessl M, Blancke Soares A, Geyer M, Dehmelt L, Nalbant P. FHOD1 regulates stress fiber organization by controlling the dynamics of transverse arcs and dorsal fibers. J Cell Sci 2014; 127:1379-93. [PMID: 24481812 DOI: 10.1242/jcs.134627] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The formin FHOD1 (formin homology 2 domain containing protein 1) can act as a capping and bundling protein in vitro. In cells, active FHOD1 stimulates the formation of ventral stress fibers. However, the cellular mechanisms by which this phenotype is produced and the physiological relevance of FHOD1 function are not currently understood. Here, we first show that FHOD1 controls the formation of two distinct stress fiber precursors differentially. On the one hand, it inhibits dorsal fiber growth, which requires the polymerization of parallel bundles of long actin filaments. On the other hand, it stimulates transverse arcs that are formed by the fusion of short antiparallel actin filaments. This combined action is crucial for the maturation of stress fibers and their spatio-temporal organization, and a lack of FHOD1 function perturbs dynamic cell behavior during cell migration. Furthermore, we show that the GTPase-binding and formin homology 3 domains (GBD and FH3) are responsible for stress fiber association and colocalization with myosin. Surprisingly, a version of FHOD1 that lacks these domains nevertheless retains its full capacity to stimulate arc and ventral stress fiber formation. Based on our findings, we propose a mechanism in which FHOD1 promotes the formation of short actin filaments and transiently associates with transverse arcs, thus providing tight temporal and spatial control of the formation and turnover of transverse arcs into mature ventral stress fibers during dynamic cell behavior.
Collapse
Affiliation(s)
- Nina Schulze
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
| | | | | | | | | | | |
Collapse
|
98
|
Bordeleau F, Alcoser TA, Reinhart-King CA. Physical biology in cancer. 5. The rocky road of metastasis: the role of cytoskeletal mechanics in cell migratory response to 3D matrix topography. Am J Physiol Cell Physiol 2014; 306:C110-20. [PMID: 24196535 PMCID: PMC3919983 DOI: 10.1152/ajpcell.00283.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 10/30/2013] [Indexed: 01/22/2023]
Abstract
The tumor microenvironment is a milieu of heterogeneous architectural features that affect tumor growth and metastatic invasion. Pore size, density, stiffness, and fiber architecture change dramatically from location to location throughout the tumor matrix. While many studies have addressed the effects of two-dimensional extracellular matrix structure and composition on cell migration, less is known about how cancer cells navigate complex, heterogeneous three-dimensional (3D) microenvironments. Mechanical structures such as actin and keratin, part of the cytoskeletal framework, and lamins, part of the nucleoskeletal framework, play a key role in migration and are altered during cancer progression. Recent evidence suggests that these changes in cytoskeletal and nucleoskeletal structures may enable cancer cells to efficiently respond to features such as pore size and stiffness to invade and migrate. Here we discuss the role of cell mechanics and the cytoskeleton in the ability of cells to navigate and respond to 3D matrix features and heterogeneities.
Collapse
Affiliation(s)
- Francois Bordeleau
- Department of Biomedical Engineering, Cornell University, Ithaca, New York
| | | | | |
Collapse
|
99
|
Peng ZM, Yu W, Xie Y, Peng WH, Cao HH, Shen JH, Wu ZY, Li EM, Xu LY. A four actin-binding protein signature model for poor prognosis of patients with esophageal squamous cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:5950-9. [PMID: 25337239 PMCID: PMC4203210 DOI: pmid/25337239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 08/20/2014] [Indexed: 02/05/2023]
Abstract
The actin cytoskeleton is a dynamic structure with actin-binding proteins (ABPs) playing an essential role in the regulation of migration, differentiation and signal transduction in all eukaryotic cells. We examined the relationship between altered expression of four ABPs and clinical parameters in esophageal squamous cell carcinoma (ESCC). To this end, we analyzed 152 formalin-fixed and paraffin-embedded esophageal curative resection specimens by immunohistochemistry for tensin, profilin-1, villin-1 and talin. A molecular predictor model, based on the combined expression of the four proteins, was developed to correlate the expression pattern of the four ABPs with clinical factors and prognosis of ESCC. According to the results, weak significance was found for tensin in lymph node metastasis (P=0.033), and profilin-1 in pTNM stage (P=0.031). However, our four-protein model showed strong correlation with the 5-year overall survival rate (P=0.002). Similarly, Kendall's tau-b test also showed the relationship between the collective expression pattern of the four ABPs with lymph node metastasis (P=0.005) and pTNM stage (P=0.001). Our results demonstrate that the collective protein expression pattern of four actin-binding proteins could be a biomarker to estimate the prognosis of ESCC patients.
Collapse
Affiliation(s)
- Zhang-Mei Peng
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical CollegeShantou 515041, P. R. China
- Institute of Oncologic Pathology, Shantou University Medical CollegeShantou 515041, P. R. China
| | - Wei Yu
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical CollegeShantou 515041, P. R. China
- Institute of Oncologic Pathology, Shantou University Medical CollegeShantou 515041, P. R. China
| | - Ying Xie
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical CollegeShantou 515041, P. R. China
- Institute of Oncologic Pathology, Shantou University Medical CollegeShantou 515041, P. R. China
| | - Wei-Hua Peng
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical CollegeShantou 515041, P. R. China
- Institute of Oncologic Pathology, Shantou University Medical CollegeShantou 515041, P. R. China
| | - Hui-Hui Cao
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical CollegeShantou 515041, P. R. China
- Institute of Oncologic Pathology, Shantou University Medical CollegeShantou 515041, P. R. China
| | - Jin-Hui Shen
- Department of Pathology, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-Sen UniversityShantou 515041, P. R. China
| | - Zhi-Yong Wu
- Department of Oncologic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-Sen UniversityShantou 515041, P. R. China
| | - En-Min Li
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical CollegeShantou 515041, P. R. China
- Department of Biochemistry and Molecular Biology, Shantou University Medical CollegeShantou 515041, P. R. China
| | - Li-Yan Xu
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical CollegeShantou 515041, P. R. China
- Institute of Oncologic Pathology, Shantou University Medical CollegeShantou 515041, P. R. China
| |
Collapse
|
100
|
Jean L, Majumdar D, Shi M, Hinkle LE, Diggins NL, Ao M, Broussard JA, Evans JC, Choma DP, Webb DJ. Activation of Rac by Asef2 promotes myosin II-dependent contractility to inhibit cell migration on type I collagen. J Cell Sci 2013; 126:5585-97. [PMID: 24144700 DOI: 10.1242/jcs.131060] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Non-muscle myosin II (MyoII) contractility is central to the regulation of numerous cellular processes, including migration. Rho is a well-characterized modulator of actomyosin contractility, but the function of other GTPases, such as Rac, in regulating contractility is currently not well understood. Here, we show that activation of Rac by the guanine nucleotide exchange factor Asef2 (also known as SPATA13) impairs migration on type I collagen through a MyoII-dependent mechanism that enhances contractility. Knockdown of endogenous Rac or treatment of cells with a Rac-specific inhibitor decreases the amount of active MyoII, as determined by serine 19 (S19) phosphorylation, and negates the Asef2-promoted increase in contractility. Moreover, treatment of cells with blebbistatin, which inhibits MyoII activity, abolishes the Asef2-mediated effect on migration. In addition, Asef2 slows the turnover of adhesions in protrusive regions of cells by promoting large mature adhesions, which has been linked to actomyosin contractility, with increased amounts of active β1 integrin. Hence, our data reveal a new role for Rac activation, promoted by Asef2, in modulating actomyosin contractility, which is important for regulating cell migration and adhesion dynamics.
Collapse
Affiliation(s)
- Léolène Jean
- Department of Biological Sciences and Vanderbilt Kennedy Center for Research on Human Development, Nashville, TN 37203, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|