301
|
Kozyrina AN, Piskova T, Di Russo J. Mechanobiology of Epithelia From the Perspective of Extracellular Matrix Heterogeneity. Front Bioeng Biotechnol 2020; 8:596599. [PMID: 33330427 PMCID: PMC7717998 DOI: 10.3389/fbioe.2020.596599] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/06/2020] [Indexed: 11/13/2022] Open
Abstract
Understanding the complexity of the extracellular matrix (ECM) and its variability is a necessary step on the way to engineering functional (bio)materials that serve their respective purposes while relying on cell adhesion. Upon adhesion, cells receive messages which contain both biochemical and mechanical information. The main focus of mechanobiology lies in investigating the role of this mechanical coordination in regulating cellular behavior. In recent years, this focus has been additionally shifted toward cell collectives and the understanding of their behavior as a whole mechanical continuum. Collective cell phenomena very much apply to epithelia which are either simple cell-sheets or more complex three-dimensional structures. Researchers have been mostly using the organization of monolayers to observe their collective behavior in well-defined experimental setups in vitro. Nevertheless, recent studies have also reported the impact of ECM remodeling on epithelial morphogenesis in vivo. These new concepts, combined with the knowledge of ECM biochemical complexity are of key importance for engineering new interactive materials to support both epithelial remodeling and homeostasis. In this review, we summarize the structure and heterogeneity of the ECM before discussing its impact on the epithelial mechanobiology.
Collapse
Affiliation(s)
- Aleksandra N. Kozyrina
- Interdisciplinary Centre for Clinical Research, RWTH Aachen University, Aachen, Germany
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Aachen, Germany
| | - Teodora Piskova
- Interdisciplinary Centre for Clinical Research, RWTH Aachen University, Aachen, Germany
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Aachen, Germany
| | - Jacopo Di Russo
- Interdisciplinary Centre for Clinical Research, RWTH Aachen University, Aachen, Germany
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Aachen, Germany
- DWI – Leibniz-Institute for Interactive Materials, Aachen, Germany
| |
Collapse
|
302
|
Insulin/Glucose-Responsive Cells Derived from Induced Pluripotent Stem Cells: Disease Modeling and Treatment of Diabetes. Cells 2020; 9:cells9112465. [PMID: 33198288 PMCID: PMC7696367 DOI: 10.3390/cells9112465] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/03/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetes, characterized by dysfunction of pancreatic β-cells and insulin resistance in peripheral organs, accounts for more than 90% of all diabetes. Despite current developments of new drugs and strategies to prevent/treat diabetes, there is no ideal therapy targeting all aspects of the disease. Restoration, however, of insulin-producing β-cells, as well as insulin-responsive cells, would be a logical strategy for the treatment of diabetes. In recent years, generation of transplantable cells derived from stem cells in vitro has emerged as an important research area. Pluripotent stem cells, either embryonic or induced, are alternative and feasible sources of insulin-secreting and glucose-responsive cells. This notwithstanding, consistent generation of robust glucose/insulin-responsive cells remains challenging. In this review, we describe basic concepts of the generation of induced pluripotent stem cells and subsequent differentiation of these into pancreatic β-like cells, myotubes, as well as adipocyte- and hepatocyte-like cells. Use of these for modeling of human disease is now feasible, while development of replacement therapies requires continued efforts.
Collapse
|
303
|
Rajendan AK, Arisaka Y, Yui N, Iseki S. Polyrotaxanes as emerging biomaterials for tissue engineering applications: a brief review. Inflamm Regen 2020; 40:27. [PMID: 33292785 PMCID: PMC7657355 DOI: 10.1186/s41232-020-00136-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023] Open
Abstract
The field of tissue engineering and regeneration constantly explores the possibility of utilizing various biomaterials' properties to achieve effective and uneventful tissue repairs. Polyrotaxanes (PRXs) are supramolecular assemblies, which possess interesting mechanical property at a molecular scale termed as molecular mobility. This molecular mobility could be utilized to stimulate various cellular mechanosignaling elements, thereby altering the cellular functions. Apart from this, the versatile nature of PRXs such as the ability to form complex with growth factors and peptides, numerous sites for chemical modifications, and processability into different forms makes them interesting candidates for applications towards tissue engineering. This literature briefly reviews the concepts of PRXs and molecular mobility, the versatile nature of PRXs, and its emerging utility towards certain tissue engineering applications.
Collapse
Affiliation(s)
- Arun Kumar Rajendan
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Yoshinori Arisaka
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo, 101-0062, Japan
| | - Nobuhiko Yui
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo, 101-0062, Japan
| | - Sachiko Iseki
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan.
| |
Collapse
|
304
|
Saffioti NA, Cavalcanti-Adam EA, Pallarola D. Biosensors for Studies on Adhesion-Mediated Cellular Responses to Their Microenvironment. Front Bioeng Biotechnol 2020; 8:597950. [PMID: 33262979 PMCID: PMC7685988 DOI: 10.3389/fbioe.2020.597950] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 10/12/2020] [Indexed: 12/28/2022] Open
Abstract
Cells interact with their microenvironment by constantly sensing mechanical and chemical cues converting them into biochemical signals. These processes allow cells to respond and adapt to changes in their environment, and are crucial for most cellular functions. Understanding the mechanism underlying this complex interplay at the cell-matrix interface is of fundamental value to decipher key biochemical and mechanical factors regulating cell fate. The combination of material science and surface chemistry aided in the creation of controllable environments to study cell mechanosensing and mechanotransduction. Biologically inspired materials tailored with specific bioactive molecules, desired physical properties and tunable topography have emerged as suitable tools to study cell behavior. Among these materials, synthetic cell interfaces with built-in sensing capabilities are highly advantageous to measure biophysical and biochemical interaction between cells and their environment. In this review, we discuss the design of micro and nanostructured biomaterials engineered not only to mimic the structure, properties, and function of the cellular microenvironment, but also to obtain quantitative information on how cells sense and probe specific adhesive cues from the extracellular domain. This type of responsive biointerfaces provides a readout of mechanics, biochemistry, and electrical activity in real time allowing observation of cellular processes with molecular specificity. Specifically designed sensors based on advanced optical and electrochemical readout are discussed. We further provide an insight into the emerging role of multifunctional micro and nanosensors to control and monitor cell functions by means of material design.
Collapse
Affiliation(s)
- Nicolás Andrés Saffioti
- Instituto de Nanosistemas, Universidad Nacional de General San Martín, San Martín, Argentina
| | | | - Diego Pallarola
- Instituto de Nanosistemas, Universidad Nacional de General San Martín, San Martín, Argentina
| |
Collapse
|
305
|
Schaeske J, Fadeeva E, Schlie-Wolter S, Deiwick A, Chichkov BN, Ingendoh-Tsakmakidis A, Stiesch M, Winkel A. Cell Type-Specific Adhesion and Migration on Laser-Structured Opaque Surfaces. Int J Mol Sci 2020; 21:ijms21228442. [PMID: 33182746 PMCID: PMC7696563 DOI: 10.3390/ijms21228442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 11/16/2022] Open
Abstract
Cytocompatibility is essential for implant approval. However, initial in vitro screenings mainly include the quantity of adherent immortalized cells and cytotoxicity. Other vital parameters, such as cell migration and an in-depth understanding of the interaction between native tissue cells and implant surfaces, are rarely considered. We investigated different laser-fabricated spike structures using primary and immortalized cell lines of fibroblasts and osteoblasts and included quantification of the cell area, aspect ratio, and focal adhesions. Furthermore, we examined the three-dimensional cell interactions with spike topographies and developed a tailored migration assay for long-term monitoring on opaque materials. While fibroblasts and osteoblasts on small spikes retained their normal morphology, cells on medium and large spikes sank into the structures, affecting the composition of the cytoskeleton and thereby changing cell shape. Up to 14 days, migration appeared stronger on small spikes, probably as a consequence of adequate focal adhesion formation and an intact cytoskeleton, whereas human primary cells revealed differences in comparison to immortalized cell lines. The use of primary cells, analysis of the cell-implant structure interaction as well as cell migration might strengthen the evaluation of cytocompatibility and thereby improve the validity regarding the putative in vivo performance of implant material.
Collapse
Affiliation(s)
- Jörn Schaeske
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany; (J.S.); (A.I.-T.); (M.S.)
| | - Elena Fadeeva
- Institute of Quantum Optics, Leibniz University of Hannover, Welfengarten 1, 30167 Hannover, Germany; (E.F.); (S.S.-W.); (A.D.); (B.N.C.)
| | - Sabrina Schlie-Wolter
- Institute of Quantum Optics, Leibniz University of Hannover, Welfengarten 1, 30167 Hannover, Germany; (E.F.); (S.S.-W.); (A.D.); (B.N.C.)
| | - Andrea Deiwick
- Institute of Quantum Optics, Leibniz University of Hannover, Welfengarten 1, 30167 Hannover, Germany; (E.F.); (S.S.-W.); (A.D.); (B.N.C.)
| | - Boris N. Chichkov
- Institute of Quantum Optics, Leibniz University of Hannover, Welfengarten 1, 30167 Hannover, Germany; (E.F.); (S.S.-W.); (A.D.); (B.N.C.)
| | - Alexandra Ingendoh-Tsakmakidis
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany; (J.S.); (A.I.-T.); (M.S.)
| | - Meike Stiesch
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany; (J.S.); (A.I.-T.); (M.S.)
| | - Andreas Winkel
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany; (J.S.); (A.I.-T.); (M.S.)
- Correspondence:
| |
Collapse
|
306
|
Malvar S, Cardoso LOB, Karolski B, Perpetuo EA, Carmo BS, Meneghini JR. A rheological approach to identify efficient biopolymer producing bacteria. Biotechnol Bioeng 2020; 118:622-632. [PMID: 33090455 DOI: 10.1002/bit.27595] [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: 06/12/2020] [Revised: 09/22/2020] [Accepted: 09/28/2020] [Indexed: 11/09/2022]
Abstract
This study investigates the relationship between collective motion and propulsion of bacterial consortia and their biopolymer production efficiency. Rheological tests were conducted for suspensions of two different methanotrophic bacterial consortia obtained after enrichment of sediment samples from mangrove sites in Brazil. We considered the linear viscoelasticity region and analyzed the values of storage and loss moduli as functions of days of cultivation, for different values of the volume fraction. The suspensions' rheological behaviors reflected the bacterial growth stage. We found that the formation of structures over time in some types of consortia can hinder the movement of bacteria in the search for nutrients. The change in complex viscosity of the two consortia followed a different and rich behavior that appears to be closely related to their capacity to capture methane. Our analysis showed a possible correlation between collective motion, viscosity reduction, and biopolymer production. The pieces of evidence from this study suggest that the efficiency of bacterial motion is directly related to biopolymer production, and this could facilitate the process of identifying the best consortium of biopolymer producing bacteria.
Collapse
Affiliation(s)
- Sara Malvar
- Department of Mechanical Engineering, Escola Politécnica, University of São Paulo, São Paulo, SP, Brazil
| | - Letícia O B Cardoso
- The Interunit Program in Biotechnology, University of São Paulo, São Paulo, SP, Brazil.,Environmental Research and Education Center - CEPEMA, Escola Politécnica, University of São Paulo, Cubatão, SP, Brazil
| | - Bruno Karolski
- Environmental Research and Education Center - CEPEMA, Escola Politécnica, University of São Paulo, Cubatão, SP, Brazil
| | - Elen A Perpetuo
- Environmental Research and Education Center - CEPEMA, Escola Politécnica, University of São Paulo, Cubatão, SP, Brazil.,Institute of Marine Sciences, Federal University of São Paulo, Santos, SP, Brazil
| | - Bruno S Carmo
- Department of Mechanical Engineering, Escola Politécnica, University of São Paulo, São Paulo, SP, Brazil
| | - Julio R Meneghini
- Department of Mechanical Engineering, Escola Politécnica, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
307
|
Shatkin G, Yeoman B, Birmingham K, Katira P, Engler AJ. Computational models of migration modes improve our understanding of metastasis. APL Bioeng 2020; 4:041505. [PMID: 33195959 PMCID: PMC7647620 DOI: 10.1063/5.0023748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/23/2020] [Indexed: 01/07/2023] Open
Abstract
Tumor cells migrate through changing microenvironments of diseased and healthy tissue, making their migration particularly challenging to describe. To better understand this process, computational models have been developed for both the ameboid and mesenchymal modes of cell migration. Here, we review various approaches that have been used to account for the physical environment's effect on cell migration in computational models, with a focus on their application to understanding cancer metastasis and the related phenomenon of durotaxis. We then discuss how mesenchymal migration models typically simulate complex cell–extracellular matrix (ECM) interactions, while ameboid migration models use a cell-focused approach that largely ignores ECM when not acting as a physical barrier. This approach greatly simplifies or ignores the mechanosensing ability of ameboid migrating cells and should be reevaluated in future models. We conclude by describing future model elements that have not been included to date but would enhance model accuracy.
Collapse
Affiliation(s)
- Gabriel Shatkin
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, USA
| | | | - Katherine Birmingham
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, USA
| | | | | |
Collapse
|
308
|
DeCamp SJ, Tsuda VMK, Ferruzzi J, Koehler SA, Giblin JT, Roblyer D, Zaman MH, Weiss ST, Kılıç A, De Marzio M, Park CY, Ogassavara NC, Mitchel JA, Butler JP, Fredberg JJ. Epithelial layer unjamming shifts energy metabolism toward glycolysis. Sci Rep 2020; 10:18302. [PMID: 33110128 PMCID: PMC7591531 DOI: 10.1038/s41598-020-74992-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/29/2020] [Indexed: 01/06/2023] Open
Abstract
In development of an embryo, healing of a wound, or progression of a carcinoma, a requisite event is collective epithelial cellular migration. For example, cells at the advancing front of a wound edge tend to migrate collectively, elongate substantially, and exert tractions more forcefully compared with cells many ranks behind. With regards to energy metabolism, striking spatial gradients have recently been reported in the wounded epithelium, as well as in the tumor, but within the wounded cell layer little is known about the link between mechanical events and underlying energy metabolism. Using the advancing confluent monolayer of MDCKII cells as a model system, here we report at single cell resolution the evolving spatiotemporal fields of cell migration speeds, cell shapes, and traction forces measured simultaneously with fields of multiple indices of cellular energy metabolism. Compared with the epithelial layer that is unwounded, which is non-migratory, solid-like and jammed, the leading edge of the advancing cell layer is shown to become progressively more migratory, fluid-like, and unjammed. In doing so the cytoplasmic redox ratio becomes progressively smaller, the NADH lifetime becomes progressively shorter, and the mitochondrial membrane potential and glucose uptake become progressively larger. These observations indicate that a metabolic shift toward glycolysis accompanies collective cellular migration but show, further, that this shift occurs throughout the cell layer, even in regions where associated changes in cell shapes, traction forces, and migration velocities have yet to penetrate. In characterizing the wound healing process these morphological, mechanical, and metabolic observations, taken on a cell-by-cell basis, comprise the most comprehensive set of biophysical data yet reported. Together, these data suggest the novel hypothesis that the unjammed phase evolved to accommodate fluid-like migratory dynamics during episodes of tissue wound healing, development, and plasticity, but is more energetically expensive compared with the jammed phase, which evolved to maintain a solid-like non-migratory state that is more energetically economical.
Collapse
Affiliation(s)
- Stephen J DeCamp
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Victor M K Tsuda
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Jacopo Ferruzzi
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Stephan A Koehler
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - John T Giblin
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Darren Roblyer
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Muhammad H Zaman
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Howard Hughes Medical Institute, Boston University, Boston, MA, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ayşe Kılıç
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Margherita De Marzio
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Chan Young Park
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Nicolas Chiu Ogassavara
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Jennifer A Mitchel
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - James P Butler
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jeffrey J Fredberg
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
309
|
Mechanically tuning actin filaments to modulate the action of actin-binding proteins. Curr Opin Cell Biol 2020; 68:72-80. [PMID: 33160108 DOI: 10.1016/j.ceb.2020.09.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 12/16/2022]
Abstract
In cells, the actin cytoskeleton is regulated by an interplay between mechanics and biochemistry. A key mechanism, which has emerged based on converging indications from structural, cellular, and biophysical data, depicts the actin filament as a mechanically tunable substrate: mechanical stress applied to an actin filament induces conformational changes, which modify the binding and the regulatory action of actin-binding proteins. For a long time, however, direct evidence of this mechanotransductive mechanism was very scarce. This situation is changing rapidly, and recent in vitro single-filament studies using different techniques have revealed that several actin-binding proteins are able to sense tension, curvature, and/or torsion, applied to actin filaments. Here, we discuss these recent advances and their possible implications.
Collapse
|
310
|
Cells into tubes: Molecular and physical principles underlying lumen formation in tubular organs. Curr Top Dev Biol 2020; 143:37-74. [PMID: 33820625 DOI: 10.1016/bs.ctdb.2020.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tubular networks, such as the vascular and respiratory systems, transport liquids and gases in multicellular organisms. The basic units of these organs are tubes formed by single or multiple cells enclosing a luminal cavity. The formation and maintenance of correctly sized and shaped lumina are fundamental steps in organogenesis and are essential for organismal homeostasis. Therefore, understanding how cells generate, shape and maintain lumina is crucial for understanding normal organogenesis as well as the basis of pathological conditions. Lumen formation involves polarized membrane trafficking, cytoskeletal dynamics, and the influence of intracellular as well as extracellular mechanical forces, such as cortical tension, luminal pressure or blood flow. Various tissue culture and in vivo model systems, ranging from MDCK cell spheroids to tubular organs in worms, flies, fish, and mice, have provided many insights into the molecular and cellular mechanisms underlying lumenogenesis and revealed key factors that regulate the size and shape of cellular tubes. Moreover, the development of new experimental and imaging approaches enabled quantitative analyses of intracellular dynamics and allowed to assess the roles of cellular and tissue mechanics during tubulogenesis. However, how intracellular processes are coordinated and regulated across scales of biological organization to generate properly sized and shaped tubes is only beginning to be understood. Here, we review recent insights into the molecular, cellular and physical mechanisms underlying lumen formation during organogenesis. We discuss how these mechanisms control lumen formation in various model systems, with a special focus on the morphogenesis of tubular organs in Drosophila.
Collapse
|
311
|
Pratt SJP, Lee RM, Chang KT, Hernández-Ochoa EO, Annis DA, Ory EC, Thompson KN, Bailey PC, Mathias TJ, Ju JA, Vitolo MI, Schneider MF, Stains JP, Ward CW, Martin SS. Mechanoactivation of NOX2-generated ROS elicits persistent TRPM8 Ca 2+ signals that are inhibited by oncogenic KRas. Proc Natl Acad Sci U S A 2020; 117:26008-26019. [PMID: 33020304 PMCID: PMC7584994 DOI: 10.1073/pnas.2009495117] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Changes in the mechanical microenvironment and mechanical signals are observed during tumor progression, malignant transformation, and metastasis. In this context, understanding the molecular details of mechanotransduction signaling may provide unique therapeutic targets. Here, we report that normal breast epithelial cells are mechanically sensitive, responding to transient mechanical stimuli through a two-part calcium signaling mechanism. We observed an immediate, robust rise in intracellular calcium (within seconds) followed by a persistent extracellular calcium influx (up to 30 min). This persistent calcium was sustained via microtubule-dependent mechanoactivation of NADPH oxidase 2 (NOX2)-generated reactive oxygen species (ROS), which acted on transient receptor potential cation channel subfamily M member 8 (TRPM8) channels to prolong calcium signaling. In contrast, the introduction of a constitutively active oncogenic KRas mutation inhibited the magnitude of initial calcium signaling and severely blunted persistent calcium influx. The identification that oncogenic KRas suppresses mechanically-induced calcium at the level of ROS provides a mechanism for how KRas could alter cell responses to tumor microenvironment mechanics and may reveal chemotherapeutic targets for cancer. Moreover, we find that expression changes in both NOX2 and TRPM8 mRNA predict poor clinical outcome in estrogen receptor (ER)-negative breast cancer patients, a population with limited available treatment options. The clinical and mechanistic data demonstrating disruption of this mechanically-activated calcium pathway in breast cancer patients and by KRas activation reveal signaling alterations that could influence cancer cell responses to the tumor mechanical microenvironment and impact patient survival.
Collapse
Affiliation(s)
- Stephen J P Pratt
- Program in Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201;
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Rachel M Lee
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Katarina T Chang
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Erick O Hernández-Ochoa
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - David A Annis
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Eleanor C Ory
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Keyata N Thompson
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Patrick C Bailey
- Program in Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Trevor J Mathias
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Julia A Ju
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Michele I Vitolo
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Martin F Schneider
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Joseph P Stains
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Christopher W Ward
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201
- School of Nursing, University of Maryland, Baltimore, MD 21201
| | - Stuart S Martin
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201;
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| |
Collapse
|
312
|
Tran VD, Kumar S. Transduction of cell and matrix geometric cues by the actin cytoskeleton. Curr Opin Cell Biol 2020; 68:64-71. [PMID: 33075689 DOI: 10.1016/j.ceb.2020.08.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022]
Abstract
Engineered culture substrates have proven invaluable for investigating the role of cell and extracellular matrix geometry in governing cell behavior. While the mechanisms relating geometry to phenotype are complex, it is clear that the actin cytoskeleton plays a key role in integrating geometric inputs and transducing these cues into intracellular signals that drive downstream biology. Here, we review recent progress in elucidating the role of the cell and matrix geometry in regulating actin cytoskeletal architecture and mechanics. We address new developments in traditional two-dimensional culture paradigms and discuss efforts to extend these advances to three-dimensional systems, ranging from nanotextured surfaces to microtopographical systems (e.g. channels) to fully three-dimensional matrices.
Collapse
Affiliation(s)
- Vivien D Tran
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, USA
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, USA.
| |
Collapse
|
313
|
Garcia MA, Sadeghipour E, Engel L, Nelson WJ, Pruitt BL. MEMS device for applying shear and tension to an epithelium combined with fluorescent live cell imaging. JOURNAL OF MICROMECHANICS AND MICROENGINEERING : STRUCTURES, DEVICES, AND SYSTEMS 2020; 30:125004. [PMID: 34413578 PMCID: PMC8372846 DOI: 10.1088/1361-6439/abb12c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Mechanical forces play important roles in the biological function of cells and tissues. While numerous studies have probed the force response of cells and measured cell-generated forces, they have primarily focused on tensile, but not shear forces. Here, we describe the design, fabrication, and application of a silicon micromachined device that is capable of independently applying and sensing both tensile and shear forces in an epithelial cell monolayer. We integrated the device with an upright microscope to enable live cell brightfield and fluorescent imaging of cells over many hours following mechanical perturbation. Using devices of increasing stiffness and the same displacement input, we demonstrate that epithelia exhibit concomitant higher maximum resistive tensile forces and quicker force relaxation. In addition, we characterized the force response of the epithelium to cyclic shear loading. While the maximum resistive forces of epithelia under cyclic shear perturbation remained unchanged between cycles, cyclic loading led to faster relaxation of the resistive forces. The device presented here can be applied to studying the force response of other monolayer-forming cell types and is compatible with pharmacological perturbation of cell structures and functions.
Collapse
Affiliation(s)
- Miguel A Garcia
- Department of Biology, Stanford University, Stanford, CA 94305, United States of America
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, United States of America
| | - Ehsan Sadeghipour
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States of America
- Department of Bioengineering, Stanford University, Stanford, CA 94305, United States of America
| | - Leeya Engel
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States of America
- Department of Bioengineering, Stanford University, Stanford, CA 94305, United States of America
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - W James Nelson
- Department of Biology, Stanford University, Stanford, CA 94305, United States of America
| | - Beth L Pruitt
- Department of Mechanical Engineering, University of California, Santa Barbara, CA 93106, United States of America
- Department of Biomolecular Science and Engineering, University of California, Santa Barbara, CA 93106, United States of America
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, United States of America
| |
Collapse
|
314
|
Díaz-Díaz C, Baonza G, Martín-Belmonte F. The vertebrate epithelial apical junctional complex: Dynamic interplay between Rho GTPase activity and cell polarization processes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183398. [DOI: 10.1016/j.bbamem.2020.183398] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 12/31/2022]
|
315
|
Farahat M, Kazi GAS, Hara ES, Matsumoto T. Effect of Biomechanical Environment on Degeneration of Meckel's Cartilage. J Dent Res 2020; 100:171-178. [PMID: 33000980 DOI: 10.1177/0022034520960118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
During orofacial tissue development, the anterior and posterior regions of the Meckel's cartilage undergo mineralization, while the middle region undergoes degeneration. Despite the interesting and particular phenomena, the mechanisms that regulate the different fates of Meckel's cartilage, including the effects of biomechanical cues, are still unclear. Therefore, the purpose of this study was to systematically investigate the course of Meckel's cartilage during embryonic development from a biomechanical perspective. Histomorphological and biomechanical (stiffness) changes in the Meckel's cartilage were analyzed from embryonic day 12 to postnatal day 0. The results revealed remarkable changes in the morphology and size of chondrocytes, as well as the occurrence of chondrocyte burst in the vicinity of the mineralization site, an often-seen phenomenon preceding endochondral ossification. To understand the effect of biomechanical cues on Meckel's cartilage fate, a mechanically tuned 3-dimensional hydrogel culture system was used. At the anterior region, a moderately soft environment (10-kPa hydrogel) promoted chondrocyte burst and ossification. On the contrary, at the middle region, a more rigid environment (40-kPa hydrogel) enhanced cartilage degradation by inducing a higher expression of MMP-1 and MMP-13. These results indicate that differences in the biomechanical properties of the surrounding environment are essential factors that distinctly guide the mineralization and degradation of Meckel's cartilage and would be valuable tools for modulating in vitro cartilage and bone tissue engineering.
Collapse
Affiliation(s)
- M Farahat
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - G A S Kazi
- Department of Applied Life Systems Engineering, Graduate School of Science and Engineering, Yamagata University, Yamagata, Japan
| | - E S Hara
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - T Matsumoto
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
316
|
Ouyang M, Qian Z, Bu B, Jin Y, Wang J, Zhu Y, Liu L, Pan Y, Deng L. Sensing Traction Force on the Matrix Induces Cell-Cell Distant Mechanical Communications for Self-Assembly. ACS Biomater Sci Eng 2020; 6:5833-5848. [PMID: 33320570 DOI: 10.1021/acsbiomaterials.0c01035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The long-range biomechanical force propagating across a large scale may reserve the capability to trigger coordinative responses within cell population such as during angiogenesis, epithelial tubulogenesis, and cancer metastasis. How cells communicate in a distant manner within the group for self-assembly remains largely unknown. Here, we found that airway smooth muscle cells (ASMCs) rapidly self-assembled into a well-constructed network on 3D Matrigel containing type I collagen (COL), which relied on long-range biomechanical force across the matrix to direct cell-cell distant interactions. Similar results happened by HUVEC cells to mimic angiogenesis. Interestingly, single ASMCs initiated multiple extended protrusions precisely pointing to neighboring cells in distance (100-300 μm away or 5-10 folds of the diameter of a round single cell), depending on traction force sensing. Individual ASMCs mechanosensed each other to move directionally on both nonfibrous Matrigel only and Matrigel containing fibrous COL but lost mutual sensing on the cross-linked gel or coated glass due to no long-range force transmission. The bead tracking assay demonstrated distant transmission of traction force (up to 400 μm) during the matrix deformation, and finite element method modeling confirmed the consistency between maximum strain distribution on the matrix and cell directional movements in experiments. Furthermore, ASMCs recruited COL from the hydrogel to build a fibrous network to mechanically stabilize the cell network. Our results revealed principally that cells can sense traction force transmitted through the matrix to initiate cell-cell distant mechanical communications, resulting in cell directional migration and coordinated cell and COL self-assembly with active matrix remodeling. As an interesting phenomenon, cells seem to be able to "make a phone call" via long-range biomechanics, which implicates physiological importance such as for tissue pattern formation.
Collapse
Affiliation(s)
- Mingxing Ouyang
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Zhili Qian
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Bing Bu
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Yang Jin
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Jiajia Wang
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Yiming Zhu
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Lei Liu
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Yan Pan
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Linhong Deng
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| |
Collapse
|
317
|
Fedele C, Mäntylä E, Belardi B, Hamkins-Indik T, Cavalli S, Netti PA, Fletcher DA, Nymark S, Priimagi A, Ihalainen TO. Azobenzene-based sinusoidal surface topography drives focal adhesion confinement and guides collective migration of epithelial cells. Sci Rep 2020; 10:15329. [PMID: 32948792 PMCID: PMC7501301 DOI: 10.1038/s41598-020-71567-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 08/14/2020] [Indexed: 01/09/2023] Open
Abstract
Surface topography is a key parameter in regulating the morphology and behavior of single cells. At multicellular level, coordinated cell displacements drive many biological events such as embryonic morphogenesis. However, the effect of surface topography on collective migration of epithelium has not been studied in detail. Mastering the connection between surface features and collective cellular behaviour is highly important for novel approaches in tissue engineering and repair. Herein, we used photopatterned microtopographies on azobenzene-containing materials and showed that smooth topographical cues with proper period and orientation can efficiently orchestrate cell alignment in growing epithelium. Furthermore, the experimental system allowed us to investigate how the orientation of the topographical features can alter the speed of wound closure in vitro. Our findings indicate that the extracellular microenvironment topography coordinates their focal adhesion distribution and alignment. These topographic cues are able to guide the collective migration of multicellular systems, even when cell-cell junctions are disrupted.
Collapse
Affiliation(s)
- Chiara Fedele
- Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland
| | - Elina Mäntylä
- BioMediTech and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Brian Belardi
- Department of Bioengineering and Biophysics Program, University of California, Berkeley, CA, 94720, USA
| | - Tiama Hamkins-Indik
- Department of Bioengineering and Biophysics Program, University of California, Berkeley, CA, 94720, USA
| | - Silvia Cavalli
- Istituto Italiano Di Tecnologia, Center for Advanced Biomaterials for Healthcare @CRIB, Naples, Italy
| | - Paolo A Netti
- Istituto Italiano Di Tecnologia, Center for Advanced Biomaterials for Healthcare @CRIB, Naples, Italy
| | - Daniel A Fletcher
- Department of Bioengineering and Biophysics Program, University of California, Berkeley, CA, 94720, USA
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Soile Nymark
- BioMediTech and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Arri Priimagi
- Faculty of Engineering and Natural Sciences, Tampere University, Tampere, Finland.
| | - Teemu O Ihalainen
- BioMediTech and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
318
|
Tian F, Lin TC, Wang L, Chen S, Chen X, Yiu PM, Tsui OKC, Chu J, Kiang CH, Park H. Mechanical Responses of Breast Cancer Cells to Substrates of Varying Stiffness Revealed by Single-Cell Measurements. J Phys Chem Lett 2020; 11:7643-7649. [PMID: 32794712 DOI: 10.1021/acs.jpclett.0c02065] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
How cancer cells respond to different mechanical environments remains elusive. Here, we investigated the tension in single focal adhesions of MDA-MB-231 (metastatic breast cancer cells) and MCF-10A (normal human breast cells) cells on substrates of varying stiffness using single-cell measurements. Tension measurements in single focal adhesions using an improved FRET-based tension sensor showed that the tension in focal adhesions of MDA-MB-231 cells increased on stiffer substrates while the tension in MCF-10A cells exhibited no apparent change against the substrate stiffness. Viscoelasticity measurements using magnetic tweezers showed that the power-law exponent of MDA-MB-231 cells decreased on stiffer substrates whereas MCF-10A cells had similar exponents throughout the whole stiffness, indicating that MDA-MB-231 cells change their viscoelasticity on stiffer substrates. Such changes in tension in focal adhesions and viscoelasticity against the substrate stiffness represent an adaptability of cancer cells in mechanical environments, which can facilitate the metastasis of cancer cells to different tissues.
Collapse
Affiliation(s)
- Fang Tian
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
| | - Tsung-Cheng Lin
- Department of Physics & Astronomy, Rice University, Houston, Texas 77005, United States
| | - Liang Wang
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Sidong Chen
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
| | - Xingxiang Chen
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
| | - Pak Man Yiu
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
| | - Ophelia K C Tsui
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
| | - Jun Chu
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ching-Hwa Kiang
- Department of Physics & Astronomy, Rice University, Houston, Texas 77005, United States
| | - Hyokeun Park
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
| |
Collapse
|
319
|
CONNECTOME or COLLECTOME? A NEUROPHILOSOPHICAL Perspective. Integr Psychol Behav Sci 2020; 56:266-279. [PMID: 32939700 DOI: 10.1007/s12124-020-09576-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2020] [Indexed: 11/27/2022]
Abstract
Human beings exist in a biological and social system from a micro to a macro level, by means of "collectivity", a dynamic collaboration that they have established together with the elements in that system in a way to complement each other and realize a common goal. Many neuroscientific concepts used today to explain neuronal processes from which mental functions originate are far from searching answers to traditional philosophical questions. However, the brain - as the generator of highly abstract concepts - is so complex that it cannot be explained by minimalistic approaches. The concept of connectome used in recent years to describe neuronal connections from which brain functions originate exemplifies this minimalistic approach, because it only describes structural and functional connections but does not look at brain functions in a holistic view. For this reason, we propose the concept of collectome - to replace the concept of connectome - that describes a homeomorphic and homotopic neuronal framework that has a bicontinuous style of work from micro to macroscale which is based on fractal rules.
Collapse
|
320
|
Buenzli PR, Lanaro M, Wong CS, McLaughlin MP, Allenby MC, Woodruff MA, Simpson MJ. Cell proliferation and migration explain pore bridging dynamics in 3D printed scaffolds of different pore size. Acta Biomater 2020; 114:285-295. [PMID: 32673750 DOI: 10.1016/j.actbio.2020.07.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/11/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023]
Abstract
Tissue growth in bioscaffolds is influenced significantly by pore geometry, but how this geometric dependence emerges from dynamic cellular processes such as cell proliferation and cell migration remains poorly understood. Here we investigate the influence of pore size on the time required to bridge pores in thin 3D-printed scaffolds. Experimentally, new tissue infills the pores continually from their perimeter under strong curvature control, which leads the tissue front to round off with time. Despite the varied shapes assumed by the tissue during this evolution, we find that time to bridge a pore simply increases linearly with the overall pore size. To disentangle the biological influence of cell behaviour and the mechanistic influence of geometry in this experimental observation, we propose a simple reaction-diffusion model of tissue growth based on Porous-Fisher invasion of cells into the pores. First, this model provides a good qualitative representation of the evolution of the tissue; new tissue in the model grows at an effective rate that depends on the local curvature of the tissue substrate. Second, the model suggests that a linear dependence of bridging time with pore size arises due to geometric reasons alone, not to differences in cell behaviours across pores of different sizes. Our analysis suggests that tissue growth dynamics in these experimental constructs is dominated by mechanistic crowding effects that influence collective cell proliferation and migration processes, and that can be predicted by simple reaction-diffusion models of cells that have robust, consistent behaviours.
Collapse
Affiliation(s)
- Pascal R Buenzli
- School of Mathematical Sciences, Queensland University of Technology (QUT), Brisbane, Australia.
| | - Matthew Lanaro
- School of Mechanical Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, Australia
| | - Cynthia S Wong
- School of Mechanical Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, Australia
| | - Maximilian P McLaughlin
- School of Mechanical Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, Australia
| | - Mark C Allenby
- School of Mechanical Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, Australia
| | - Maria A Woodruff
- School of Mechanical Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, Australia
| | - Matthew J Simpson
- School of Mathematical Sciences, Queensland University of Technology (QUT), Brisbane, Australia
| |
Collapse
|
321
|
Xu XP, Pokutta S, Torres M, Swift MF, Hanein D, Volkmann N, Weis WI. Structural basis of αE-catenin-F-actin catch bond behavior. eLife 2020; 9:e60878. [PMID: 32915141 PMCID: PMC7588230 DOI: 10.7554/elife.60878] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/09/2020] [Indexed: 11/13/2022] Open
Abstract
Cell-cell and cell-matrix junctions transmit mechanical forces during tissue morphogenesis and homeostasis. α-Catenin links cell-cell adhesion complexes to the actin cytoskeleton, and mechanical load strengthens its binding to F-actin in a direction-sensitive manner. Specifically, optical trap experiments revealed that force promotes a transition between weak and strong actin-bound states. Here, we describe the cryo-electron microscopy structure of the F-actin-bound αE-catenin actin-binding domain, which in solution forms a five-helix bundle. In the actin-bound structure, the first helix of the bundle dissociates and the remaining four helices and connecting loops rearrange to form the interface with actin. Deletion of the first helix produces strong actin binding in the absence of force, suggesting that the actin-bound structure corresponds to the strong state. Our analysis explains how mechanical force applied to αE-catenin or its homolog vinculin favors the strongly bound state, and the dependence of catch bond strength on the direction of applied force.
Collapse
Affiliation(s)
| | - Sabine Pokutta
- Departments of Structural Biology and Molecular & Cellular Physiology, Stanford University School of MedicineStanfordUnited States
| | - Megan Torres
- Departments of Structural Biology and Molecular & Cellular Physiology, Stanford University School of MedicineStanfordUnited States
| | | | - Dorit Hanein
- Scintillon InstituteSan DiegoUnited States
- Department of Structural Biology and Chemistry, Pasteur InstituteParisFrance
| | - Niels Volkmann
- Scintillon InstituteSan DiegoUnited States
- Department of Structural Biology and Chemistry, Pasteur InstituteParisFrance
| | - William I Weis
- Departments of Structural Biology and Molecular & Cellular Physiology, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
322
|
Song M, Kim J, Shin H, Kim Y, Jang H, Park Y, Kim SJ. Development of Magnetic Torque Stimulation (MTS) Utilizing Rotating Uniform Magnetic Field for Mechanical Activation of Cardiac Cells. NANOMATERIALS 2020; 10:nano10091684. [PMID: 32867131 PMCID: PMC7557977 DOI: 10.3390/nano10091684] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 12/13/2022]
Abstract
Regulation of cell signaling through physical stimulation is an emerging topic in biomedicine. Background: While recent advances in biophysical technologies show capabilities for spatiotemporal stimulation, interfacing those tools with biological systems for intact signal transfer and noncontact stimulation remains challenging. Here, we describe the use of a magnetic torque stimulation (MTS) system combined with engineered magnetic particles to apply forces on the surface of individual cells. MTS utilizes an externally rotating magnetic field to induce a spin on magnetic particles and generate torsional force to stimulate mechanotransduction pathways in two types of human heart cells—cardiomyocytes and cardiac fibroblasts. Methods: The MTS system operates in a noncontact mode with two magnets separated (60 mm) from each other and generates a torque of up to 15 pN µm across the entire area of a 35-mm cell culture dish. The MTS system can mechanically stimulate both types of human heart cells, inducing maturation and hypertrophy. Results: Our findings show that application of the MTS system under hypoxic conditions induces not only nuclear localization of mechanoresponsive YAP proteins in human heart cells but also overexpression of hypertrophy markers, including β-myosin heavy chain (βMHC), cardiotrophin-1 (CT-1), microRNA-21 (miR-21), and transforming growth factor beta-1 (TGFβ-1). Conclusions: These results have important implications for the applicability of the MTS system to diverse in vitro studies that require remote and noninvasive mechanical regulation.
Collapse
Affiliation(s)
- Myeongjin Song
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; (M.S.); (J.K.); (Y.K.); (H.J.)
| | - Jongseong Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; (M.S.); (J.K.); (Y.K.); (H.J.)
| | - Hyundo Shin
- Department of Mechanical Engineering, Yonsei University, Seoul 03722, Korea;
| | - Yekwang Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; (M.S.); (J.K.); (Y.K.); (H.J.)
| | - Hwanseok Jang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; (M.S.); (J.K.); (Y.K.); (H.J.)
| | - Yongdoo Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; (M.S.); (J.K.); (Y.K.); (H.J.)
- Correspondence: (Y.P.); (S.-J.K.); Tel.: +82-2-2286-1460 (Y.P.)
| | - Seung-Jong Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; (M.S.); (J.K.); (Y.K.); (H.J.)
- Correspondence: (Y.P.); (S.-J.K.); Tel.: +82-2-2286-1460 (Y.P.)
| |
Collapse
|
323
|
Antagonistic Functions of Connexin 43 during the Development of Primary or Secondary Bone Tumors. Biomolecules 2020; 10:biom10091240. [PMID: 32859065 PMCID: PMC7565206 DOI: 10.3390/biom10091240] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022] Open
Abstract
Despite research and clinical advances during recent decades, bone cancers remain a leading cause of death worldwide. There is a low survival rate for patients with primary bone tumors such as osteosarcoma and Ewing’s sarcoma or secondary bone tumors such as bone metastases from prostate carcinoma. Gap junctions are specialized plasma membrane structures consisting of transmembrane channels that directly link the cytoplasm of adjacent cells, thereby enabling the direct exchange of small signaling molecules between cells. Discoveries of human genetic disorders due to genetic mutations in gap junction proteins (connexins) and experimental data using connexin knockout mice have provided significant evidence that gap-junctional intercellular communication (Gj) is crucial for tissue function. Thus, the dysfunction of Gj may be responsible for the development of some diseases. Gj is thus a main mechanism for tumor cells to communicate with other tumor cells and their surrounding microenvironment to survive and proliferate. If it is well accepted that a low level of connexin expression favors cancer cell proliferation and therefore primary tumor development, more evidence is suggesting that a high level of connexin expression stimulates various cellular process such as intravasation, extravasation, or migration of metastatic cells. If so, connexin expression would facilitate secondary tumor dissemination. This paper discusses evidence that suggests that connexin 43 plays an antagonistic role in the development of primary bone tumors as a tumor suppressor and secondary bone tumors as a tumor promoter.
Collapse
|
324
|
Liu Q, Zheng S, Ye K, He J, Shen Y, Cui S, Huang J, Gu Y, Ding J. Cell migration regulated by RGD nanospacing and enhanced under moderate cell adhesion on biomaterials. Biomaterials 2020; 263:120327. [PMID: 32927304 DOI: 10.1016/j.biomaterials.2020.120327] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/21/2022]
Abstract
While nanoscale modification of a biomaterial surface is known to influence various cell behaviors, it is unclear whether there is an optimal nanospacing of a bioactive ligand with respect to cell migration. Herein, we investigated the effects of nanospacing of arginine-glycine-aspartate (RGD) peptide on cell migration and its relation to cell adhesion. To this end, we prepared RGD nanopatterns with varied nanospacings (31-125 nm) against the nonfouling background of poly(ethylene glycol), and employed human umbilical vein endothelial cells (HUVECs) to examine cell behaviors on the nanopatterned surfaces. While HUVECs adhered well on surfaces of RGD nanospacing less than 70 nm and exhibited a monotonic decrease of adhesion with the increase of RGD nanospacing, cell migration exhibited a nonmonotonic change with the ligand nanospacing: the maximum migration velocity was observed around 90 nm of nanospacing, and slow or very slow migration occurred in the cases of small or large RGD nanospacings. Therefore, moderate cell adhesion is beneficial for fast cell migration. Further molecular biology studies revealed that attenuated cell adhesion and activated dynamic actin rearrangement accounted for the promotion of cell migration, and the genes of small G proteins such as Cdc42 were upregulated correspondingly. The present study sheds new light on cell migration and its relation to cell adhesion, and paves a way for designing biomaterials for applications in regenerative medicine.
Collapse
Affiliation(s)
- Qiong Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China; Navy Special Medical Center, The Second Military Medical University, Shanghai, 200433, China
| | - Shuang Zheng
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Kai Ye
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Junhao He
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Yang Shen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Shuquan Cui
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Jiale Huang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Yexin Gu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, China; Zhuhai Fudan Innovation Institute, Zhuhai, Guangdong, 519000, China.
| |
Collapse
|
325
|
Moura CC, Miranda A, Oreffo ROC, De Beule PAA. Correlative fluorescence and atomic force microscopy to advance the bio-physical characterisation of co-culture of living cells. Biochem Biophys Res Commun 2020; 529:392-397. [PMID: 32703441 DOI: 10.1016/j.bbrc.2020.06.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 11/29/2022]
Abstract
An understanding of the cell mechanical properties involved in numerous cellular processes including cell division, cell migration/invasion, and cell morphology, is crucial in developing and informing cell physiology and function. Atomic force microscopy (AFM) offers a powerful biophysical technique that facilitates the imaging of living cells under physiological buffer conditions. However, AFM in isolation cannot discriminate between different cell types within heterogeneous samples for example in a solid biopsy. The current studies demonstrate the potential of AFM in combination with correlative fluorescence optical sectioning microscopy for live cell imaging. Furthermore, this work establishes the advantage of fluorescence-AFM imaging to distinguish and analyse single-cell bio-physical properties in mixed human cell populations, in real-time. Critically, our results show that correlative fluorescence-AFM imaging allows the simultaneous co-localised detection of fluorescence coupled with nano-mechanical mapping. The findings from this work contribute to the promotion and dissemination of correlative multimodal imaging in life sciences, providing a platform for further investigations in biological and pre-clinical research.
Collapse
Affiliation(s)
- Catarina Costa Moura
- INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, 4715-330, Braga, Portugal.
| | - Adelaide Miranda
- INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, 4715-330, Braga, Portugal
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - Pieter A A De Beule
- INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, 4715-330, Braga, Portugal
| |
Collapse
|
326
|
Zhang G, Mueller R, Doostmohammadi A, Yeomans JM. Active inter-cellular forces in collective cell motility. J R Soc Interface 2020; 17:20200312. [PMID: 32781933 DOI: 10.1098/rsif.2020.0312] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The collective behaviour of confluent cell sheets is strongly influenced both by polar forces, arising through cytoskeletal propulsion, and by active inter-cellular forces, which are mediated by interactions across cell-cell junctions. We use a phase-field model to explore the interplay between these two contributions and compare the dynamics of a cell sheet when the polarity of the cells aligns to (i) their main axis of elongation, (ii) their velocity and (iii) when the polarity direction executes a persistent random walk. In all three cases, we observe a sharp transition from a jammed state (where cell rearrangements are strongly suppressed) to a liquid state (where the cells can move freely relative to each other) when either the polar or the inter-cellular forces are increased. In addition, for case (ii) only, we observe an additional dynamical state, flocking (solid or liquid), where the majority of the cells move in the same direction. The flocking state is seen for strong polar forces, but is destroyed as the strength of the inter-cellular activity is increased.
Collapse
Affiliation(s)
- Guanming Zhang
- The Rudolf Peierls Centre for Theoretical Physics, University of Oxford, Parks Road, Oxford OX1 3PU, UK
| | - Romain Mueller
- The Rudolf Peierls Centre for Theoretical Physics, University of Oxford, Parks Road, Oxford OX1 3PU, UK
| | - Amin Doostmohammadi
- The Niels Bohr Institute, University of Copenhagen, Blegdamsvej 17, 2100 Copenhagen, DK
| | - Julia M Yeomans
- The Rudolf Peierls Centre for Theoretical Physics, University of Oxford, Parks Road, Oxford OX1 3PU, UK
| |
Collapse
|
327
|
Liu Z, Wang L, Xu H, Du Q, Li L, Wang L, Zhang ES, Chen G, Wang Y. Heterogeneous Responses to Mechanical Force of Prostate Cancer Cells Inducing Different Metastasis Patterns. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903583. [PMID: 32775149 PMCID: PMC7404165 DOI: 10.1002/advs.201903583] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/10/2020] [Indexed: 05/15/2023]
Abstract
The physical cues in the extracellular environment play important roles in cancer cell metastasis. However, how metastatic cancer cells respond to the diverse mechanical environments of metastatic sites is not fully understood. Here, substrates with different mechanical properties are prepared to simulate the extracellular mechanical environment of various human tissues. The prostate cancer (PC) cells derived from different cancer metastasis sites show heterogeneity in mechanical response. This heterogeneity mediates two distinct metastasis patterns. High stiffness promotes individual cell migration and proliferation by inducing Yes-associated protein and tafazzin (YAP/TAZ) nuclear localization in bone metastasis-derived cells, whereas low stiffness promotes cell migration and proliferation by inducing lymphatic metastasis-derived cells to form clusters characterized by high expression of CD44. The different metastasis patterns induced by the mechanical properties of the extracellular environment are crucial in the development of PC.
Collapse
Affiliation(s)
- Zhixiao Liu
- Research Center of Developmental BiologyDepartment of Histology and EmbryologyCollege of Basic MedicineSecond Military Medical UniversityShanghai200433China
- Department of UrologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620China
| | - Liujun Wang
- Research Center of Developmental BiologyDepartment of Histology and EmbryologyCollege of Basic MedicineSecond Military Medical UniversityShanghai200433China
| | - Huan Xu
- Department of UrologyChanghai Hospital of ShanghaiSecond Military Medical UniversityShanghai200433China
- Department of PathologyDuke University School of MedicineDurhamNC27710USA
| | - Qiqige Du
- The State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular ScienceFudan UniversityShanghai200433China
| | - Li Li
- Research Center of Developmental BiologyDepartment of Histology and EmbryologyCollege of Basic MedicineSecond Military Medical UniversityShanghai200433China
| | - Ling Wang
- Stem Cell and Regeneration Medicine InstituteResearch Center of Translational MedicineSecond Military Medical UniversityShanghai200433China
- Shanghai Key Laboratory of Cell EngineeringSecond Military Medical UniversityShanghai200433China
| | - En Song Zhang
- The State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular ScienceFudan UniversityShanghai200433China
| | - Guosong Chen
- The State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular ScienceFudan UniversityShanghai200433China
| | - Yue Wang
- Research Center of Developmental BiologyDepartment of Histology and EmbryologyCollege of Basic MedicineSecond Military Medical UniversityShanghai200433China
- Stem Cell and Regeneration Medicine InstituteResearch Center of Translational MedicineSecond Military Medical UniversityShanghai200433China
- Shanghai Key Laboratory of Cell EngineeringSecond Military Medical UniversityShanghai200433China
| |
Collapse
|
328
|
Lohmann S, Giampietro C, Pramotton FM, Al‐Nuaimi D, Poli A, Maiuri P, Poulikakos D, Ferrari A. The Role of Tricellulin in Epithelial Jamming and Unjamming via Segmentation of Tricellular Junctions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001213. [PMID: 32775171 PMCID: PMC7404176 DOI: 10.1002/advs.202001213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/18/2020] [Indexed: 06/11/2023]
Abstract
Collective cellular behavior in confluent monolayers supports physiological and pathological processes of epithelial development, regeneration, and carcinogenesis. Here, the attainment of a mature and static tissue configuration or the local reactivation of cell motility involve a dynamic regulation of the junctions established between neighboring cells. Tricellular junctions (tTJs), established at vertexes where three cells meet, are ideally located to control cellular shape and coordinate multicellular movements. However, their function in epithelial tissue dynamic remains poorly defined. To investigate the role of tTJs establishment and maturation in the jamming and unjamming transitions of epithelial monolayers, a semi-automatic image-processing pipeline is developed and validated enabling the unbiased and spatially resolved determination of the tTJ maturity state based on the localization of fluorescent reporters. The software resolves the variation of tTJ maturity accompanying collective transitions during tissue maturation, wound healing, and upon the adaptation to osmolarity changes. Altogether, this work establishes junctional maturity at tricellular contacts as a novel biological descriptor of collective responses in epithelial monolayers.
Collapse
Affiliation(s)
- Sophie Lohmann
- Laboratory of Thermodynamics in Emerging TechnologiesETH ZurichZurich8092Switzerland
| | - Costanza Giampietro
- EMPASwiss Federal Laboratories for Materials Science and TechnologyExperimental Continuum MechanicsDübendorf8600Switzerland
| | | | - Dunja Al‐Nuaimi
- Laboratory of Thermodynamics in Emerging TechnologiesETH ZurichZurich8092Switzerland
| | - Alessandro Poli
- IFOM‐ The FIRC Institute of Molecular OncologySpatiotemporal organization of the nucleus UnitMilan20139Italy
| | - Paolo Maiuri
- IFOM‐ The FIRC Institute of Molecular OncologySpatiotemporal organization of the nucleus UnitMilan20139Italy
| | - Dimos Poulikakos
- Laboratory of Thermodynamics in Emerging TechnologiesETH ZurichZurich8092Switzerland
| | - Aldo Ferrari
- Laboratory of Thermodynamics in Emerging TechnologiesETH ZurichZurich8092Switzerland
- EMPASwiss Federal Laboratories for Materials Science and TechnologyExperimental Continuum MechanicsDübendorf8600Switzerland
- Institute for Mechanical SystemsETH ZurichZürich8092Switzerland
| |
Collapse
|
329
|
Zhang B, Zhu M, Li Z, Lung PS, Chrzanowski W, Kwok CT, Lu J, Li Q. Cellular fate of deformable needle-shaped PLGA-PEG fibers. Acta Biomater 2020; 112:182-189. [PMID: 32470525 DOI: 10.1016/j.actbio.2020.05.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
Abstract
Deformability of micro/nanometer sized particles plays an important role in particle-cell interactions and thus becomes a key parameter in carrier design in biomedicine application such as drug delivery and vaccinology. Yet the influence of material's deformability on the cellular fate of the particles as well as physiology response of live cells are to be understood. Here we show the cellular fate of needle shaped (high aspect ratio ~25) PLGA-PEG copolymer fibers depending on their deformability. We found that all the fibers entered murine macrophage cells (RAW 264.7) via phagocytosis. While the fibers of high apparent Young's modulus (average value = 872 kPa) maintained their original shape upon phagocytosis, their counterparts of low apparent Young's modulus (average value = 56 kPa) curled in cells. The observed deformation of fibers of low apparent Young's modulus in cells coincided with abnormal intracellular actin translocation and absence of lysosome/phagosome fusion in macrophages, suggesting the important role of material mechanical properties and mechano-related cellular pathway in affecting cell physiology. STATEMENT OF SIGNIFICANCE: Particles are increasingly important in the field of biomedicine, especially when they are serving as drug carriers. Physical cues, such as mechanical properties, were shown to provide insight into their stability and influence on physiology inside the cell. In the current study, we managed to fabricate 5 types of needle shaped PLGA-PEG fibers with controlled Young's modulus. We found that hard fibers maintained their original shape upon phagocytosis, while soft fibers were curled by actin compressive force inside the cell, causing abnormal actin translocation and impediment of lysosome/phagosome fusion, suggesting the important role of material mechanical properties and mechano-related cellular pathway in affecting cell physiology.
Collapse
|
330
|
|
331
|
Massou S, Nunes Vicente F, Wetzel F, Mehidi A, Strehle D, Leduc C, Voituriez R, Rossier O, Nassoy P, Giannone G. Cell stretching is amplified by active actin remodelling to deform and recruit proteins in mechanosensitive structures. Nat Cell Biol 2020; 22:1011-1023. [PMID: 32719553 DOI: 10.1038/s41556-020-0548-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 06/22/2020] [Indexed: 02/07/2023]
Abstract
Detection and conversion of mechanical forces into biochemical signals controls cell functions during physiological and pathological processes. Mechanosensing is based on protein deformations and reorganizations, yet the molecular mechanisms are still unclear. Using a cell-stretching device compatible with super-resolution microscopy and single-protein tracking, we explored the nanoscale deformations and reorganizations of individual proteins inside mechanosensitive structures. We achieved super-resolution microscopy after live stretching on intermediate filaments, microtubules and integrin adhesions. Simultaneous single-protein tracking and stretching showed that while integrins followed the elastic deformation of the substrate, actin filaments and talin also displayed lagged and transient inelastic responses associated with active acto-myosin remodelling and talin deformations. Capturing acute reorganizations of single molecules during stretching showed that force-dependent vinculin recruitment is delayed and depends on the maturation of integrin adhesions. Thus, cells respond to external forces by amplifying transiently and locally cytoskeleton displacements, enabling protein deformation and recruitment in mechanosensitive structures.
Collapse
Affiliation(s)
- Sophie Massou
- Interdisciplinary Institute for Neuroscience, UMR 5297, Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, UMR 5297, CNRS, Bordeaux, France
| | - Filipe Nunes Vicente
- Interdisciplinary Institute for Neuroscience, UMR 5297, Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, UMR 5297, CNRS, Bordeaux, France
| | - Franziska Wetzel
- Laboratoire Photonique Numérique et Nanosciences, UMR 5298, Université de Bordeaux, Talence, France.,Laboratoire Photonique Numérique et Nanosciences, UMR 5298, Institut d'Optique and CNRS, Talence, France
| | - Amine Mehidi
- Interdisciplinary Institute for Neuroscience, UMR 5297, Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, UMR 5297, CNRS, Bordeaux, France
| | - Dan Strehle
- Laboratoire Photonique Numérique et Nanosciences, UMR 5298, Université de Bordeaux, Talence, France.,Laboratoire Photonique Numérique et Nanosciences, UMR 5298, Institut d'Optique and CNRS, Talence, France
| | - Cecile Leduc
- Cell Polarity, Migration and Cancer Unit, UMR 3691, Institut Pasteur Paris and CNRS, Paris, France
| | - Raphaël Voituriez
- Laboratoire Jean Perrin and Laboratoire de Physique Théorique de la Matière Condensée, CNRS - Sorbonne Université, Paris, France
| | - Olivier Rossier
- Interdisciplinary Institute for Neuroscience, UMR 5297, Université de Bordeaux, Bordeaux, France.,Interdisciplinary Institute for Neuroscience, UMR 5297, CNRS, Bordeaux, France
| | - Pierre Nassoy
- Laboratoire Photonique Numérique et Nanosciences, UMR 5298, Université de Bordeaux, Talence, France.,Laboratoire Photonique Numérique et Nanosciences, UMR 5298, Institut d'Optique and CNRS, Talence, France
| | - Grégory Giannone
- Interdisciplinary Institute for Neuroscience, UMR 5297, Université de Bordeaux, Bordeaux, France. .,Interdisciplinary Institute for Neuroscience, UMR 5297, CNRS, Bordeaux, France.
| |
Collapse
|
332
|
Deng Z, Maksaev G, Schlegel AM, Zhang J, Rau M, Fitzpatrick JAJ, Haswell ES, Yuan P. Structural mechanism for gating of a eukaryotic mechanosensitive channel of small conductance. Nat Commun 2020; 11:3690. [PMID: 32704140 PMCID: PMC7378837 DOI: 10.1038/s41467-020-17538-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 07/07/2020] [Indexed: 01/21/2023] Open
Abstract
Mechanosensitive ion channels transduce physical force into electrochemical signaling that underlies an array of fundamental physiological processes, including hearing, touch, proprioception, osmoregulation, and morphogenesis. The mechanosensitive channels of small conductance (MscS) constitute a remarkably diverse superfamily of channels critical for management of osmotic pressure. Here, we present cryo-electron microscopy structures of a MscS homolog from Arabidopsis thaliana, MSL1, presumably in both the closed and open states. The heptameric MSL1 channel contains an unusual bowl-shaped transmembrane region, which is reminiscent of the evolutionarily and architecturally unrelated mechanosensitive Piezo channels. Upon channel opening, the curved transmembrane domain of MSL1 flattens and expands. Our structures, in combination with functional analyses, delineate a structural mechanism by which mechanosensitive channels open under increased membrane tension. Further, the shared structural feature between unrelated channels suggests the possibility of a unified mechanical gating mechanism stemming from membrane deformation induced by a non-planar transmembrane domain. Mechanosensitive channels transduce physical force into electrochemical signaling in processes such as hearing, touch, proprioception, osmoregulation, and morphogenesis. Here, authors use cryo-electron microscopy to provide structural insights into the mechanical gating mechanism.
Collapse
Affiliation(s)
- Zengqin Deng
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA.,Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Grigory Maksaev
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA.,Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Angela M Schlegel
- Department of Biology, Washington University in Saint Louis, Saint Louis, MO, 63130, USA.,NSF Center for Engineering Mechanobiology, Washington University in Saint Louis, Saint Louis, MO, 63130, USA
| | - Jingying Zhang
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA.,Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Michael Rau
- Washington University Center for Cellular Imaging, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - James A J Fitzpatrick
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA.,Washington University Center for Cellular Imaging, Washington University School of Medicine, Saint Louis, MO, 63110, USA.,Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO, 63110, USA.,Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, 63130, USA
| | - Elizabeth S Haswell
- Department of Biology, Washington University in Saint Louis, Saint Louis, MO, 63130, USA.,NSF Center for Engineering Mechanobiology, Washington University in Saint Louis, Saint Louis, MO, 63130, USA
| | - Peng Yuan
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA. .,Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| |
Collapse
|
333
|
Zhou G, Zhang B, Wei L, Zhang H, Galluzzi M, Li J. Spatially Resolved Correlation between Stiffness Increase and Actin Aggregation around Nanofibers Internalized in Living Macrophages. MATERIALS 2020; 13:ma13143235. [PMID: 32708102 PMCID: PMC7412258 DOI: 10.3390/ma13143235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 12/21/2022]
Abstract
Plasticity and functional diversity of macrophages play an important role in resisting pathogens invasion, tumor progression and tissue repair. At present, nanodrug formulations are becoming increasingly important to induce and control the functional diversity of macrophages. In this framework, the internalization process of nanodrugs is co-regulated by a complex interplay of biochemistry, cell physiology and cell mechanics. From a biophysical perspective, little is known about cellular mechanics’ modulation induced by the nanodrug carrier’s internalization. In this study, we used the polylactic-co-glycolic acid (PLGA)–polyethylene glycol (PEG) nanofibers as a model drug carrier, and we investigated their influence on macrophage mechanics. Interestingly, the nanofibers internalized in macrophages induced a local increase of stiffness detected by atomic force microscopy (AFM) nanomechanical investigation. Confocal laser scanning microscopy revealed a thickening of actin filaments around nanofibers during the internalization process. Following geometry and mechanical properties by AFM, indentation experiments are virtualized in a finite element model simulation. It turned out that it is necessary to include an additional actin wrapping layer around nanofiber in order to achieve similar reaction force of AFM experiments, consistent with confocal observation. The quantitative investigation of actin reconfiguration around internalized nanofibers can be exploited to develop novel strategies for drug delivery.
Collapse
Affiliation(s)
- Guoqiao Zhou
- Shenzhen Key Laboratory of Nanobiomechanics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (G.Z.); (B.Z.); (L.W.); (H.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bokai Zhang
- Shenzhen Key Laboratory of Nanobiomechanics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (G.Z.); (B.Z.); (L.W.); (H.Z.)
| | - Liyu Wei
- Shenzhen Key Laboratory of Nanobiomechanics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (G.Z.); (B.Z.); (L.W.); (H.Z.)
| | - Han Zhang
- Shenzhen Key Laboratory of Nanobiomechanics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (G.Z.); (B.Z.); (L.W.); (H.Z.)
- State Key Laboratory of Traction Power, Southwest Jiaotong University, Chengdu 610000, China
| | - Massimiliano Galluzzi
- Shenzhen Key Laboratory of Nanobiomechanics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (G.Z.); (B.Z.); (L.W.); (H.Z.)
- Correspondence: (M.G.); (J.L.)
| | - Jiangyu Li
- Shenzhen Key Laboratory of Nanobiomechanics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (G.Z.); (B.Z.); (L.W.); (H.Z.)
- Correspondence: (M.G.); (J.L.)
| |
Collapse
|
334
|
Subramanianbalachandar V, Steward R. Stiffness Decouples Cellular Mechanosensation. Biophys J 2020; 119:233. [PMID: 32610091 DOI: 10.1016/j.bpj.2020.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 06/04/2020] [Indexed: 10/24/2022] Open
Affiliation(s)
| | - Robert Steward
- Departments of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, Florida; Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida.
| |
Collapse
|
335
|
Svandova E, Peterkova R, Matalova E, Lesot H. Formation and Developmental Specification of the Odontogenic and Osteogenic Mesenchymes. Front Cell Dev Biol 2020; 8:640. [PMID: 32850793 PMCID: PMC7396701 DOI: 10.3389/fcell.2020.00640] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/25/2020] [Indexed: 12/15/2022] Open
Abstract
Within the mandible, the odontogenic and osteogenic mesenchymes develop in a close proximity and form at about the same time. They both originate from the cranial neural crest. These two condensing ecto-mesenchymes are soon separated from each other by a very loose interstitial mesenchyme, whose cells do not express markers suggesting a neural crest origin. The two condensations give rise to mineralized tissues while the loose interstitial mesenchyme, remains as a soft tissue. This is crucial for proper anchorage of mammalian teeth. The situation in all three regions of the mesenchyme was compared with regard to cell heterogeneity. As the development progresses, the early phenotypic differences and the complexity in cell heterogeneity increases. The differences reported here and their evolution during development progressively specifies each of the three compartments. The aim of this review was to discuss the mechanisms underlying condensation in both the odontogenic and osteogenic compartments as well as the progressive differentiation of all three mesenchymes during development. Very early, they show physical and structural differences including cell density, shape and organization as well as the secretion of three distinct matrices, two of which will mineralize. Based on these data, this review highlights the consecutive differences in cell-cell and cell-matrix interactions, which support the cohesion as well as mechanosensing and mechanotransduction. These are involved in the conversion of mechanical energy into biochemical signals, cytoskeletal rearrangements cell differentiation, or collective cell behavior.
Collapse
Affiliation(s)
- Eva Svandova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, Academy of Sciences, Brno, Czechia
| | - Renata Peterkova
- Department of Histology and Embryology, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Eva Matalova
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, Academy of Sciences, Brno, Czechia.,Department of Physiology, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia
| | - Herve Lesot
- Laboratory of Odontogenesis and Osteogenesis, Institute of Animal Physiology and Genetics, Academy of Sciences, Brno, Czechia
| |
Collapse
|
336
|
Abuwarda H, Pathak MM. Mechanobiology of neural development. Curr Opin Cell Biol 2020; 66:104-111. [PMID: 32687993 DOI: 10.1016/j.ceb.2020.05.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/23/2020] [Accepted: 05/30/2020] [Indexed: 01/13/2023]
Abstract
As the brain develops, proliferating cells organize into structures, differentiate, migrate, extrude long processes, and connect with other cells. These biological processes produce mechanical forces that further shape cellular dynamics and organ patterning. A major unanswered question in developmental biology is how the mechanical forces produced during development are detected and transduced by cells to impact biochemical and genetic programs of development. This gap in knowledge stems from a lack of understanding of the molecular players of cellular mechanics and an absence of techniques for measuring and manipulating mechanical forces in tissue. In this review article, we examine recent advances that are beginning to clear these bottlenecks and highlight results from new approaches that reveal the role of mechanical forces in neurodevelopmental processes.
Collapse
Affiliation(s)
- Hamid Abuwarda
- Department of Physiology & Biophysics, UC Irvine, Irvine, CA, USA; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA, USA
| | - Medha M Pathak
- Department of Physiology & Biophysics, UC Irvine, Irvine, CA, USA; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA, USA; Center for Complex Biological Systems, UC Irvine, Irvine, CA, USA; Department of Biomedical Engineering, UC Irvine, Irvine, CA, USA.
| |
Collapse
|
337
|
Vishwakarma M, Spatz JP, Das T. Mechanobiology of leader-follower dynamics in epithelial cell migration. Curr Opin Cell Biol 2020; 66:97-103. [PMID: 32663734 DOI: 10.1016/j.ceb.2020.05.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/21/2022]
Abstract
Collective cell migration is fundamental to biological form and function. It is also relevant to the formation and repair of organs and to various pathological situations, including metastatic propagation of cancer. Technological, experimental, and computational advancements have allowed the researchers to explore various aspects of collective migration, spanning from biochemical signalling to inter-cellular force transduction. Here, we summarize our current understanding of the mechanobiology of collective cell migration, limiting to epithelial tissues. On the basis of recent studies, we describe how cells sense and respond to guidance signals to orchestrate various modes of migration and identify the determining factors dictating leader-follower interactions. We highlight how the inherent mechanics of dense epithelial monolayers at multicellular length scale might instruct individual cells to behave collectively. On the basis of these findings, we propose that mechanical resilience, obtained by a certain extent of cell jamming, allows the epithelium to perform efficient collective migration during wound healing.
Collapse
Affiliation(s)
- Medhavi Vishwakarma
- School of Cellular and Molecular Medicine, University of Bristol, University Walk, Bristol BS81TD, United Kingdom; Department of Cellular Biophysics, Max Planck Institute for Medical Research, Heidelberg 69120, Germany
| | - Joachim P Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Heidelberg 69120, Germany; Department of Biophysical Chemistry, University of Heidelberg, Heidelberg 69117, Germany
| | - Tamal Das
- TIFR Centre for Interdisciplinary Sciences, Tata Institute of Fundamental Research Hyderabad (TIFR-H), Hyderabad 500046, India.
| |
Collapse
|
338
|
Role of biomechanics in vascularization of tissue-engineered bones. J Biomech 2020; 110:109920. [PMID: 32827778 DOI: 10.1016/j.jbiomech.2020.109920] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 12/23/2022]
Abstract
Biomaterial based reconstruction is still the most commonly employed method of small bone defect reconstruction. Bone tissue-engineered techniques are improving, and adjuncts such as vascularization technologies allow re-evaluation of traditional reconstructive methods for healingofcritical-sized bone defect. Slow infiltration rate of vasculogenesis after cell-seeded scaffold implantation limits the use of clinically relevant large-sized scaffolds. Hence, in vitro vascularization within the tissue-engineered bone before implantation is required to overcome the serious challenge of low cell survival rate after implantation which affects bone tissue regeneration and osseointegration. Mechanobiological interactions between cells and microvascular mechanics regulate biological processes regarding cell behavior. In addition, load-bearing scaffolds demand mechanical stability properties after vascularization to have adequate strength while implanted. With the advent of bioreactors, vascularization has been greatly improved by biomechanical regulation of stem cell differentiation through fluid-induced shear stress and synergizing osteogenic and angiogenic differentiation in multispecies coculture cells. The benefits of vascularization are clear: avoidance of mass transfer limitation and oxygen deprivation, a significant decrease in cell necrosis, and consequently bone development, regeneration and remodeling. Here, we discuss specific techniques to avoid pitfalls and optimize vascularization results of tissue-engineered bone. Cell source, scaffold modifications and bioreactor design, and technique specifics all play a critical role in this new, and rapidly growing method for bone defect reconstruction. Given the crucial importance of long-term survival of vascular network in physiological function of 3D engineered-bone constructs, greater knowledge of vascularization approaches may lead to the development of new strategies towards stabilization of formed vascular structure.
Collapse
|
339
|
Jain N, Moeller J, Vogel V. Mechanobiology of Macrophages: How Physical Factors Coregulate Macrophage Plasticity and Phagocytosis. Annu Rev Biomed Eng 2020; 21:267-297. [PMID: 31167103 DOI: 10.1146/annurev-bioeng-062117-121224] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In addition to their early-recognized functions in host defense and the clearance of apoptotic cell debris, macrophages play vital roles in tissue development, homeostasis, and repair. If misregulated, they steer the progression of many inflammatory diseases. Much progress has been made in understanding the mechanisms underlying macrophage signaling, transcriptomics, and proteomics, under physiological and pathological conditions. Yet, the detailed mechanisms that tune circulating monocytes/macrophages and tissue-resident macrophage polarization, differentiation, specification, and their functional plasticity remain elusive. We review how physical factors affect macrophage phenotype and function, including how they hunt for particles and pathogens, as well as the implications for phagocytosis, autophagy, and polarization from proinflammatory to prohealing phenotype. We further discuss how this knowledge can be harnessed in regenerative medicine and for the design of new drugs and immune-modulatory drug delivery systems, biomaterials, and tissue scaffolds.
Collapse
Affiliation(s)
- Nikhil Jain
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, and Department of Health Sciences and Technology, ETH Zurich, CH-8093 Zurich, Switzerland;
| | - Jens Moeller
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, and Department of Health Sciences and Technology, ETH Zurich, CH-8093 Zurich, Switzerland;
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, and Department of Health Sciences and Technology, ETH Zurich, CH-8093 Zurich, Switzerland;
| |
Collapse
|
340
|
Non-Muscle Myosin 2A (NM2A): Structure, Regulation and Function. Cells 2020; 9:cells9071590. [PMID: 32630196 PMCID: PMC7408548 DOI: 10.3390/cells9071590] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/30/2022] Open
Abstract
Non-muscle myosin 2A (NM2A) is a motor cytoskeletal enzyme with crucial importance from the early stages of development until adulthood. Due to its capacity to convert chemical energy into force, NM2A powers the contraction of the actomyosin cytoskeleton, required for proper cell division, adhesion and migration, among other cellular functions. Although NM2A has been extensively studied, new findings revealed that a lot remains to be discovered concerning its spatiotemporal regulation in the intracellular environment. In recent years, new functions were attributed to NM2A and its activity was associated to a plethora of illnesses, including neurological disorders and infectious diseases. Here, we provide a concise overview on the current knowledge regarding the structure, the function and the regulation of NM2A. In addition, we recapitulate NM2A-associated diseases and discuss its potential as a therapeutic target.
Collapse
|
341
|
Jain S, Cachoux VM, Narayana GH, de Beco S, D’Alessandro J, Cellerin V, Chen T, Heuzé ML, Marcq P, Mège RM, Kabla AJ, Lim CT, Ladoux B. The role of single cell mechanical behavior and polarity in driving collective cell migration. NATURE PHYSICS 2020; 16:802-809. [PMID: 32641972 PMCID: PMC7343533 DOI: 10.1038/s41567-020-0875-z] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/11/2020] [Indexed: 05/19/2023]
Abstract
The directed migration of cell collectives is essential in various physiological processes, such as epiboly, intestinal epithelial turnover, and convergent extension during morphogenesis as well as during pathological events like wound healing and cancer metastasis. Collective cell migration leads to the emergence of coordinated movements over multiple cells. Our current understanding emphasizes that these movements are mainly driven by large-scale transmission of signals through adherens junctions. In this study, we show that collective movements of epithelial cells can be triggered by polarity signals at the single cell level through the establishment of coordinated lamellipodial protrusions. We designed a minimalistic model system to generate one-dimensional epithelial trains confined in ring shaped patterns that recapitulate rotational movements observed in vitro in cellular monolayers and in vivo in genitalia or follicular cell rotation. Using our system, we demonstrated that cells follow coordinated rotational movements after the establishment of directed Rac1-dependent polarity over the entire monolayer. Our experimental and numerical approaches show that the maintenance of coordinated migration requires the acquisition of a front-back polarity within each single cell but does not require the maintenance of cell-cell junctions. Taken together, these unexpected findings demonstrate that collective cell dynamics in closed environments as observed in multiple in vitro and in vivo situations can arise from single cell behavior through a sustained memory of cell polarity.
Collapse
Affiliation(s)
- Shreyansh Jain
- Institut Jacques Monod, CNRS UMR 7592, Université de Paris, Paris 75013, France
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | | | | | - Simon de Beco
- Institut Jacques Monod, CNRS UMR 7592, Université de Paris, Paris 75013, France
| | - Joseph D’Alessandro
- Institut Jacques Monod, CNRS UMR 7592, Université de Paris, Paris 75013, France
| | - Victor Cellerin
- Institut Jacques Monod, CNRS UMR 7592, Université de Paris, Paris 75013, France
| | - Tianchi Chen
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Mélina L. Heuzé
- Institut Jacques Monod, CNRS UMR 7592, Université de Paris, Paris 75013, France
| | - Philippe Marcq
- PMMH, CNRS, ESPCI Paris, PSL University, Sorbonne Université, Université de Paris, F-75005, Paris, France
| | - René-Marc Mège
- Institut Jacques Monod, CNRS UMR 7592, Université de Paris, Paris 75013, France
| | - Alexandre J. Kabla
- Engineering Department, University of Cambridge, Cambridge CB2 1PZ, United Kingdom
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Division of Biomedical Engineering, 4 Engineering Drive 3, National University of Singapore, Singapore 117583
- Institute for Health Innovation & Technology (iHealthtech), National University of Singapore, MD6, 14 Medical Drive, Singapore 117599
| | - Benoit Ladoux
- Institut Jacques Monod, CNRS UMR 7592, Université de Paris, Paris 75013, France
- Correspondence to:
| |
Collapse
|
342
|
The Mechanical Microenvironment in Breast Cancer. Cancers (Basel) 2020; 12:cancers12061452. [PMID: 32503141 PMCID: PMC7352870 DOI: 10.3390/cancers12061452] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/27/2020] [Accepted: 06/01/2020] [Indexed: 01/22/2023] Open
Abstract
Mechanotransduction is the interpretation of physical cues by cells through mechanosensation mechanisms that elegantly translate mechanical stimuli into biochemical signaling pathways. While mechanical stress and their resulting cellular responses occur in normal physiologic contexts, there are a variety of cancer-associated physical cues present in the tumor microenvironment that are pathological in breast cancer. Mechanistic in vitro data and in vivo evidence currently support three mechanical stressors as mechanical modifiers in breast cancer that will be the focus of this review: stiffness, interstitial fluid pressure, and solid stress. Increases in stiffness, interstitial fluid pressure, and solid stress are thought to promote malignant phenotypes in normal breast epithelial cells, as well as exacerbate malignant phenotypes in breast cancer cells.
Collapse
|
343
|
Beauchesne CC, Chabanon M, Smaniotto B, Ladoux B, Goyeau B, David B. Channeling Effect and Tissue Morphology in a Perfusion Bioreactor Imaged by X-Ray Microtomography. Tissue Eng Regen Med 2020; 17:301-311. [PMID: 32314312 PMCID: PMC7260345 DOI: 10.1007/s13770-020-00246-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Perfusion bioreactors for tissue engineering hold great promises. Indeed, the perfusion of culture medium enhances species transport and mechanically stimulates the cells, thereby increasing cell proliferation and tissue formation. Nonetheless, their development is still hampered by a lack of understanding of the relationship between mechanical cues and tissue growth. METHODS Combining tissue engineering, three-dimensional visualization and numerical simulations, we analyze the morphological evolution of neo-tissue in a model bioreactor with respect to the local flow pattern. NIH-3T3 cells were grown under perfusion for one, two and three weeks on a stack of 2 mm polyacetal beads. The model bioreactor was then imaged by X-ray micro-tomography and local tissue morphology was analyzed. To relate experimental observations and mechanical stimulii, a computational fluid dynamics model of flow around spheres in a canal was developed and solved using the finite element method. RESULTS We observe a preferential tissue formation at the bioreactor periphery, and relate it to a channeling effect leading to regions of higher flow intensity. Additionally, we find that circular crater-like tissue patterns form in narrow channel regions at early culture times. Using computational fluid dynamic simulations, we show that the location and morphology of these patterns match those of shear stress maxima. Finally, the morphology of the tissue is qualitatively described as the tissue grows and reorganizes itself. CONCLUSION Altogether, our study points out the key role of local flow conditions on the tissue morphology developed on a stack of beads in perfusion bioreactors and provides new insights for effective design of hydrodynamic bioreactors for tissue engineering using bead packings.
Collapse
Affiliation(s)
- Claire C Beauchesne
- Lab. EM2C, UPR CNRS 288, CentraleSupélec, Université Paris-Saclay, 3 rue Joliot-Curie, 91192, Gif-sur-Yvette Cedex, France
- Lab. MSSMat, UMR CNRS 8579, CentraleSupélec, Université Paris-Saclay, 3 rue Joliot-Curie, 91192, Gif-sur-Yvette Cedex, France
| | - Morgan Chabanon
- Single Molecule Biophotonics Lab. ICFO, The Institute of Photonic Sciences, av. Carl Friedrich Gauss, 3, 08860, Castelldefels, Barcelona, Spain
| | - Benjamin Smaniotto
- ENS Paris Saclay, LMT, CNRS, UMR 8535, 61 avenue du Président Wilson, 94230, Cachan, France
| | - Benoît Ladoux
- Institut Jacques Monod (IJM), UMR CNRS 7592, Université Paris Diderot, 15 rue Hélène Brion, 75013, Paris, France
| | - Benoît Goyeau
- Lab. EM2C, UPR CNRS 288, CentraleSupélec, Université Paris-Saclay, 3 rue Joliot-Curie, 91192, Gif-sur-Yvette Cedex, France.
| | - Bertrand David
- Lab. MSSMat, UMR CNRS 8579, CentraleSupélec, Université Paris-Saclay, 3 rue Joliot-Curie, 91192, Gif-sur-Yvette Cedex, France
| |
Collapse
|
344
|
Delon LC, Nilghaz A, Cheah E, Prestidge C, Thierry B. Unlocking the Potential of Organ-on-Chip Models through Pumpless and Tubeless Microfluidics. Adv Healthc Mater 2020; 9:e1901784. [PMID: 32342669 DOI: 10.1002/adhm.201901784] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/05/2020] [Indexed: 12/27/2022]
Abstract
Microfluidic organs-on-chips are rapidly being developed toward eliminating the shortcomings of static in vitro models and better addressing basic and translational research questions. A critical aspect is the dynamic culture environment they provide. However, the associated inherent requirement for controlled fluid shear stress (FSS) and therefore the need for precise pumps limits their implementation. To address this issue, here a novel approach to manufacture pumpless and tubeless organs-on-chips is reported. It relies on the use of a hydrophilic thread to provide a driving force for the perfusion of the cell culture medium through constant evaporation in the controlled conditions of a cell incubator. Well-defined and tuneable flow rates can be applied by adjusting the length and/or diameter of the thread. This approach for the preparation of an intestine-on-chip model based on the Caco-2 cell line is validated. Five days culture under 0.02 dyn·cm-2 shear conditions yield monolayers similar to those prepared using a high-precision peristaltic pump. A pumpless device can also be used to delineate the effect of FSS on the phenotype of adenocarcinomic human alveolar basal epithelial A549 cells. It is anticipated that the pumpless approach will facilitate and herefore increase the use of organs-on-chips models in the future.
Collapse
Affiliation(s)
- Ludivine C. Delon
- Future Industries Institute and ARC Centre of Excellence Convergent Bio‐Nano Science and Technology University of South Australia Mawson Lakes Campus Adelaide SA 5095 Australia
- School of Pharmacy and Medical Sciences and ARC Centre of Excellence Convergent Bio‐Nano Science and Technology University of South Australia City West Campus Adelaide SA 5000 Australia
| | - Azadeh Nilghaz
- Future Industries Institute and ARC Centre of Excellence Convergent Bio‐Nano Science and Technology University of South Australia Mawson Lakes Campus Adelaide SA 5095 Australia
| | - Edward Cheah
- Future Industries Institute and ARC Centre of Excellence Convergent Bio‐Nano Science and Technology University of South Australia Mawson Lakes Campus Adelaide SA 5095 Australia
| | - Clive Prestidge
- School of Pharmacy and Medical Sciences and ARC Centre of Excellence Convergent Bio‐Nano Science and Technology University of South Australia City West Campus Adelaide SA 5000 Australia
| | - Benjamin Thierry
- Future Industries Institute and ARC Centre of Excellence Convergent Bio‐Nano Science and Technology University of South Australia Mawson Lakes Campus Adelaide SA 5095 Australia
- School of Pharmacy and Medical Sciences and ARC Centre of Excellence Convergent Bio‐Nano Science and Technology University of South Australia City West Campus Adelaide SA 5000 Australia
| |
Collapse
|
345
|
Luparello C, Mauro M, Lazzara V, Vazzana M. Collective Locomotion of Human Cells, Wound Healing and Their Control by Extracts and Isolated Compounds from Marine Invertebrates. Molecules 2020; 25:E2471. [PMID: 32466475 PMCID: PMC7321354 DOI: 10.3390/molecules25112471] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 02/06/2023] Open
Abstract
The collective migration of cells is a complex integrated process that represents a common theme joining morphogenesis, tissue regeneration, and tumor biology. It is known that a remarkable amount of secondary metabolites produced by aquatic invertebrates displays active pharmacological properties against a variety of diseases. The aim of this review is to pick up selected studies that report the extraction and identification of crude extracts or isolated compounds that exert a modulatory effect on collective cell locomotion and/or skin tissue reconstitution and recapitulate the molecular, biochemical, and/or physiological aspects, where available, which are associated to the substances under examination, grouping the producing species according to their taxonomic hierarchy. Taken all of the collected data into account, marine invertebrates emerge as a still poorly-exploited valuable resource of natural products that may significantly improve the process of skin regeneration and restrain tumor cell migration, as documented by in vitro and in vivo studies. Therefore, the identification of the most promising invertebrate-derived extracts/molecules for the utilization as new targets for biomedical translation merits further and more detailed investigations.
Collapse
Affiliation(s)
- Claudio Luparello
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy; (M.M.); (V.L.); (M.V.)
| | | | | | | |
Collapse
|
346
|
Zhang D, Li X, Pi C, Cai L, Liu Y, Du W, Yang W, Xie J. Osteoporosis-decreased extracellular matrix stiffness impairs connexin 43-mediated gap junction intercellular communication in osteocytes. Acta Biochim Biophys Sin (Shanghai) 2020; 52:517-526. [PMID: 32286624 DOI: 10.1093/abbs/gmaa025] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/20/2019] [Accepted: 02/24/2020] [Indexed: 02/05/2023] Open
Abstract
Osteocytes are the main sensitive and responsive cells for mechanical stimuli in bone. The connexin family enables them to communicate with each other via forming functional gap junctions. However, how osteoporosis-impaired extracellular mechanical property modulates gap junction intercellular communication in osteocytes remains elusive. In this study, we established an ovariectomy (OVX)-induced osteoporosis mouse model in vivo and a polydimethylsiloxane (PDMS)-based cell culture substrate model in vitro to explore the influence of extracellular matrix (ECM) stiffness on cell-to-cell communication in osteocytes. Firstly, we established an OVX-induced osteoporosis mouse model by characterizing the changes in radiography, morphology and histochemistry of femurs. Our results showed that osteoporosis decreased the bone matrix stiffness together with the changes including the loss of osteocytes and the decrease of protein markers. Meanwhile, the dendritic process interconnection and channel-forming protein, Cx43, were reduced in osteoporosis mice. Next we mimicked ECM stiffness changes in vitro by using PDMS substrates at ratios 1:5 for normal stiffness and 1:45 for osteoporosis stiffness. Our results showed that the decreased ECM stiffness reduced the number of dendritic processes in a single cell and gap junctions between adjacent osteocytes. We further detected the decreased expression of Cx43, in the substrate with decreased stiffness. Finally, we found that gap junction-based intercellular communication was reduced in living osteocytes in the substrate with decreased stiffness. This study demonstrates the correlation between ECM mechanical property and cell-to-cell communication in osteocytes and might pave the way for further exploration of osteoporosis in terms of biomechanics.
Collapse
Affiliation(s)
- Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Xin Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Caixia Pi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Linyi Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Yang Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Wei Du
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Wenbin Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| |
Collapse
|
347
|
Patel KD, Kim TH, Mandakhbayar N, Singh RK, Jang JH, Lee JH, Kim HW. Coating biopolymer nanofibers with carbon nanotubes accelerates tissue healing and bone regeneration through orchestrated cell- and tissue-regulatory responses. Acta Biomater 2020; 108:97-110. [PMID: 32165193 DOI: 10.1016/j.actbio.2020.03.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 02/27/2020] [Accepted: 03/04/2020] [Indexed: 02/07/2023]
Abstract
Tailoring the surface of biomaterial scaffolds has been a key strategy to modulate the cellular interactions that are helpful for tissue healing process. In particular, nanotopological surfaces have been demonstrated to regulate diverse behaviors of stem cells, such as initial adhesion, spreading and lineage specification. Here, we tailor the surface of biopolymer nanofibers with carbon nanotubes (CNTs) to create a unique bi-modal nanoscale topography (500 nm nanofiber with 25 nm nanotubes) and report the performance in modulating diverse in vivo responses including inflammation, angiogenesis, and bone regeneration. When administered to a rat subcutaneous site, the CNT-coated nanofiber exhibited significantly reduced inflammatory signs (down-regulated pro-inflammatory cytokines and macrophages gathering). Moreover, the CNT-coated nanofibers showed substantially promoted angiogenic responses, with enhanced neoblood vessel formation and angiogenic marker expression. Such stimulated tissue healing events by the CNT interfacing were evidenced in a calvarium bone defect model. The in vivo bone regeneration of the CNT- coated nanofibers was significantly accelerated, with higher bone mineral density and up-regulated osteogenic signs (OPN, OCN, BMP2) of in vivo bone forming cells. The in vitro studies using MSCs could demonstrate accelerated adhesion and osteogenic differentiation and mineralization, supporting the osteo-promoting mechanism behind the in vivo bone forming event. These findings highlight that the CNTs interfacing of biopolymer nanofibers is highly effective in reducing inflammation, promoting angiogenesis, and driving adhesion and osteogenesis of MSCs, which eventually orchestrate to accelerate tissue healing and bone regeneration process. STATEMENT OF SIGNIFICANCE: Here we demonstrate that the interfacing of biopolymer nanofibers with carbon nanotubes (CNTs) could modulate multiple interactions of cells and tissues that are ultimately helpful for the tissue healing and bone regeneration process. The CNT-coated scaffolds significantly reduced the pro-inflammatory signals while stimulating the angiogenic marker expressions. Furthermore, the CNT-coated scaffolds increased the bone matrix production of bone forming cells in vivo as well as accelerated the adhesion and osteogenic differentiation of MSCs in vitro. These collective findings highlight that the CNTs coated on the biopolymer nanofibers allow the creation of a promising platform for nanoscale engineering of biomaterial surface that can favor tissue healing and bone regeneration process, through a series of orchestrated events in anti-inflammation, pro-angiogenesis, and stem cell stimulation.
Collapse
Affiliation(s)
- Kapil D Patel
- Institue of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| | - Tae-Hyun Kim
- Institue of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Nandin Mandakhbayar
- Institue of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Rajendra K Singh
- Institue of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jun-Hyeog Jang
- Department of Biochemistry, Inha University, Incheon, Republic of Korea
| | - Jung-Hwan Lee
- Institue of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 31116, Republic of Korea; UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| | - Hae-Won Kim
- Institue of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 31116, Republic of Korea; UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea.
| |
Collapse
|
348
|
Smit HJ, Strong P. Structural Elements of the Biomechanical System of Soft Tissue. Cureus 2020; 12:e7895. [PMID: 32368430 PMCID: PMC7193180 DOI: 10.7759/cureus.7895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
In living organisms, forces are constantly generated and transmitted throughout tissue. Such forces are generated through interaction with the environment and as a result of the body’s endogenous movement. If these internally or externally originating forces exceed the ability of tissues to cope with the applied forces, (i.e. “tissue thresholds”), they will cause force-related tissue harm. However, biotensegrity systems act to prevent these forces from causing structural damage to cells and tissues. The mechanism and structure of soft tissues that enable them to maintain their integrity and prevent damage under constantly changing forces is still not fully understood. The current anatomical and physical knowledge is insufficient to assess and predict how, why, where, and when to expect force-related tissue harm. When including the concept of tensegrity and the related principles of the hierarchical organisation of the elements of the subcellular tensional homeostatic structure into current biomechanical concepts, it increases our understanding of the events in force handling in relation to the onset of force-related tissue harm: Reducing incident forces in tissue to a level that is not harmful to the involved structures is achieved by dissipation, transduction and transferring the force in multiple dimensions. To enable this, the biomechanical systems must function in a continuous and consistent way from the cellular level to the entire body to prevent local peak forces from causing harm. In this article, we explore the biomechanical system with a focus on biotensegrity concepts across several organisational levels, describing in detail how it may function and reflecting on how this might be applied to patient management.
Collapse
Affiliation(s)
- Harm Jaap Smit
- Molecular Biology, Independent Researcher, Amersfoort, NLD
| | - Phil Strong
- Independent Researcher, Oldbury-on-Severn, GBR
| |
Collapse
|
349
|
Li X, Wang J. Mechanical tumor microenvironment and transduction: cytoskeleton mediates cancer cell invasion and metastasis. Int J Biol Sci 2020; 16:2014-2028. [PMID: 32549750 PMCID: PMC7294938 DOI: 10.7150/ijbs.44943] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/15/2020] [Indexed: 12/13/2022] Open
Abstract
Metastasis is a complicated, multistep process that is responsible for over 90% of cancer-related death. Metastatic disease or the movement of cancer cells from one site to another requires dramatic remodeling of the cytoskeleton. The regulation of cancer cell migration is determined not only by biochemical factors in the microenvironment but also by the biomechanical contextual information provided by the extracellular matrix (ECM). The responses of the cytoskeleton to chemical signals are well characterized and understood. However, the mechanisms of response to mechanical signals in the form of externally applied force and forces generated by the ECM are still poorly understood. Furthermore, understanding the way cellular mechanosensors interact with the physical properties of the microenvironment and transmit the signals to activate the cytoskeletal movements may help identify an effective strategy for the treatment of cancer. Here, we will discuss the role of tumor microenvironment during cancer metastasis and how physical forces remodel the cytoskeleton through mechanosensing and transduction.
Collapse
Affiliation(s)
- Xingchen Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - Jianliu Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
- Beijing Key Laboratory of Female Pelvic Floor Disorders Diseases, Beijing, 100044, China
| |
Collapse
|
350
|
Zhou C, Zhang D, Du W, Zou J, Li X, Xie J. Substrate mechanics dictate cell-cell communication by gap junctions in stem cells from human apical papilla. Acta Biomater 2020; 107:178-193. [PMID: 32105834 DOI: 10.1016/j.actbio.2020.02.032] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/31/2020] [Accepted: 02/20/2020] [Indexed: 02/08/2023]
Abstract
It is recognized that the interaction between cells and their physical microenvironment plays a fundamental role in controlling cell behaviors and even in determining cell fate. Any change in the physical properties of the extracellular matrix (ECM), such as its topography, geometry, and stiffness, controls this interaction. In the current study, we revealed a potent interconnection between the cell-matrix interaction and cell-cell communication that is mediated by interface stiffness, and elucidated this process in stem cells from human apical papilla (hSCAPs) in terms of mechanosensing, mechanotransduction, and gap junction-mediated cell-cell communication. We first fabricated polydimethylsiloxane (PDMS) substrates with the same topography and geometry but different stiffnesses and found that the cell morphology of the hSCAPs actively changed to adapt to the difference in substrate stiffness. We also found that the hSCAPs secreted more fibronectin in response to the stiff substrate. The focal adhesion plaques were changed by altering the expression of focal adhesion kinase (FAK) and paxillin. The FAK and paxillin bound to connexin 43 and, as a result, altered the gap junction formation. By performing a Lucifer yellow transfer assay, we further confirmed that the interface stiffness mediated cell-cell communication in living hSCAPs through changes in gap junction tunnels. The intrinsic mechanism that mediated cell-cell communication by extracellular stiffness show the great influence of the interaction between cells and their external physical microenvironment and stress the importance of microenvironmental mechanics in organ development and diseases. STATEMENT OF SIGNIFICANCE: Biochemical factors could direct cell behaviors such as cell proliferation, migration, differentiation, cell cycling and apoptosis. Likewise, biophysical factors could also determine cell behaviors in all biological processes. In the current study, we revealed a potent interconnection between the cell-matrix interaction and cell-cell communication by elucidating the whole process from cell mechanosensing, mechanotransduction to gap junction-mediated cell-cell communication. This process occurs in a collective of cells but not in that of a single cell. Biophysical properties of ECM induced cell-to-cell communication indicates the importance of microenvironmental mechanics in organ development and diseases. These findings should be of great interest in all biological fields, especially in biomaterials - cell/molecular biology involved in the interactions between the cell and its matrix.
Collapse
|