1
|
Allan C, Chaudhuri O. Regulation of cell migration by extracellular matrix mechanics at a glance. J Cell Sci 2025; 138:jcs263574. [PMID: 40183462 PMCID: PMC11993253 DOI: 10.1242/jcs.263574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Cell migration occurs throughout development, tissue homeostasis and regeneration, as well as in diseases such as cancer. Cells migrate along two-dimensional (2D) surfaces or interfaces, within microtracks, or in confining three-dimensional (3D) extracellular matrices. Although the basic mechanisms of 2D migration are known, recent studies have elucidated unexpected migration behaviors associated with more complex substrates and have provided insights into their underlying molecular mechanisms. Studies using engineered biomaterials for 3D culture and microfabricated channels to replicate cell confinement observed in vivo have revealed distinct modes of migration. Across these contexts, the mechanical features of the surrounding microenvironment have emerged as major regulators of migration. In this Cell Science at a Glance article and the accompanying poster, we describe physiological contexts wherein 2D and 3D cell migration are essential, report how mechanical properties of the microenvironment regulate individual and collective cell migration, and review the mechanisms mediating these diverse modes of cell migration.
Collapse
Affiliation(s)
- Cole Allan
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, USA
- Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
2
|
Nakazawa N, Grenci G, Kameo Y, Takeda N, Sawada T, Kurisu J, Zhang Z, Toma K, Adachi T, Nonomura K, Kengaku M. PIEZO1-dependent mode switch of neuronal migration in heterogeneous microenvironments in the developing brain. Cell Rep 2025; 44:115405. [PMID: 40053456 DOI: 10.1016/j.celrep.2025.115405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 01/09/2025] [Accepted: 02/14/2025] [Indexed: 03/09/2025] Open
Abstract
The migration of newborn neurons is essential for brain morphogenesis and circuit formation, yet controversy exists regarding how neurons generate the driving force against strong mechanical stresses in crowded neural tissues. We found that cerebellar granule neurons employ a mechanosensing mechanism to switch the driving forces to maneuver in irregular brain tissue. In two-dimensional (2D) cultures, actomyosin is enriched in the leading process, exerting traction force on the cell soma. In tissue or 3D confinement, however, actomyosin concentrates at the posterior cell membrane, generating contractile forces that assist passage through narrow spaces, working alongside the traction force in the leading process. The 3D migration is initiated by the activation of a mechanosensitive channel, PIEZO1. PIEZO1-induced calcium influx in the soma triggers the PKC-ezrin cascade, which recruits actomyosin and transmits its contractile force to the posterior plasma membrane. Thus, migrating neurons adapt their motility modes in distinct extracellular environments in the developing brain.
Collapse
Affiliation(s)
- Naotaka Nakazawa
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Faculty of Science and Engineering, Kindai University, Osaka 577-8502, Japan.
| | - Gianluca Grenci
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; Biomedical Engineering Department, National University of Singapore, Singapore 117583, Singapore
| | - Yoshitaka Kameo
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan; Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8501, Japan; Graduate School of Engineering, Kyoto University, Kyoto 615-8530, Japan; College of Engineering, Shibaura Institute of Technology, Tokyo 135-8548, Japan
| | - Noriko Takeda
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Tsuyoshi Sawada
- Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8501, Japan; Graduate School of Engineering, Kyoto University, Kyoto 615-8530, Japan
| | - Junko Kurisu
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Zhejing Zhang
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Kenichi Toma
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Taiji Adachi
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan; Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8501, Japan; Graduate School of Engineering, Kyoto University, Kyoto 615-8530, Japan
| | - Keiko Nonomura
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan; Department of Life Science and Technology, Tokyo Institute of Technology, Kanagawa 226-8501, Japan; National Institute for Basic Biology, Aichi 444-8585, Japan
| | - Mineko Kengaku
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
3
|
Amitrano A, Choudhury D, Konstantopoulos K. Navigating confinement: Mechanotransduction and metabolic adaptation. Curr Opin Cell Biol 2025; 94:102487. [PMID: 39999674 DOI: 10.1016/j.ceb.2025.102487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025]
Abstract
Cell migration through confined spaces is a critical process influenced by the complex three-dimensional (3D) architecture of the local microenvironment and the surrounding extracellular matrix (ECM). Cells in vivo experience diverse fluidic signals, such as extracellular fluid viscosity, hydraulic resistance, and shear forces, as well as solid cues, like ECM stiffness and viscoelasticity. These fluidic and solid stressors activate mechanotransduction processes and regulate cell migration. They also drive metabolic reprogramming, dynamically altering glycolysis and oxidative phosphorylation to meet the cell's energy demands in different microenvironments. This review discusses recent advances on the mechanisms of cell migration in confinement and how confinement-induced cellular behavior leads to metabolic reprogramming.
Collapse
Affiliation(s)
- Alice Amitrano
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Debanik Choudhury
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA.
| |
Collapse
|
4
|
Naggay BK, Farahani SK, Gao X, Holle A, Kemkemer R. Direct current electrical fields inhibit cancer cell motility in microchannel confinements. Sci Rep 2025; 15:4605. [PMID: 39920207 PMCID: PMC11806051 DOI: 10.1038/s41598-025-87737-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
The capability of cells to sense and respond to endogenous electrical fields plays a crucial role in processes like nerve regeneration, wound healing, and development. In vitro, many cell types respond to electrical fields by migrating along the corresponding electrical field vectors. This process is known as galvano- or electrotaxis. Here we report on the combined impact of micro-confinements and direct current electrical fields (dcEFs) on the motility of MDA-MB-231 human breast cancer cells using a self-developed, easy-to-use platform with microchannels ranging from 3 μ m to 11 μ m in width and 11 μ m height. We found that MDA-MB-231 cells respond to exogenous electrical fields ranging from 100 mV mm- 1 to 1000 mV mm- 1 with altered cell motility depending on the confinement size. Our data show an overall inhibited galvanotaxis in confinements, while in contrast an enhancing effect in unconfined galvanotaxis is found. The application of direct current electrical fields to microchannels not only caused a reduction in migration speed but also decreased the number of permeating cells. By applying 1000 mV mm- 1 , single-cell permeation could be prevented in confinements of 5 μ m and smaller.
Collapse
Affiliation(s)
- Benjamin Karem Naggay
- Department of Life Sciences, Reutlingen University, 72762, Reutlingen, Germany
- Reutlingen Research Institute, Reutlingen University, 72762, Reutlingen, Germany
| | | | - Xu Gao
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117411, Singapore
| | - Andrew Holle
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117411, Singapore
| | - Ralf Kemkemer
- Department of Life Sciences, Reutlingen University, 72762, Reutlingen, Germany.
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, 69120, Heidelberg, Germany.
| |
Collapse
|
5
|
Lan BQ, Wang YJ, Yu SX, Liu W, Liu YJ. Physical effects of 3-D microenvironments on confined cell behaviors. Am J Physiol Cell Physiol 2024; 327:C1192-C1201. [PMID: 39246142 DOI: 10.1152/ajpcell.00288.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Cell migration is a fundamental and functional cellular process, influenced by a complex microenvironment consisting of different cells and extracellular matrix. Recent research has highlighted that, besides biochemical cues from the microenvironment, physical cues can also greatly alter cellular behavior. However, due to the complexity of the microenvironment, little is known about how the physical interactions between migrating cells and surrounding microenvironment instructs cell movement. Here, we explore various examples of three-dimensional microenvironment reconstruction models in vitro and describe how the physical interplay between migrating cells and the neighboring microenvironment controls cell behavior. Understanding this mechanical cooperation will provide key insights into organ development, regeneration, and tumor metastasis.
Collapse
Affiliation(s)
- Bao-Qiong Lan
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Ya-Jun Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Sai-Xi Yu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Wei Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| |
Collapse
|
6
|
Mistriotis P, Wisniewski EO, Si BR, Kalab P, Konstantopoulos K. Coordinated in confined migration: crosstalk between the nucleus and ion channel-mediated mechanosensation. Trends Cell Biol 2024; 34:809-825. [PMID: 38290913 PMCID: PMC11284253 DOI: 10.1016/j.tcb.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 12/22/2023] [Accepted: 01/05/2024] [Indexed: 02/01/2024]
Abstract
Cell surface and intracellular mechanosensors enable cells to perceive different geometric, topographical, and physical cues. Mechanosensitive ion channels (MICs) localized at the cell surface and on the nuclear envelope (NE) are among the first to sense and transduce these signals. Beyond compartmentalizing the genome of the cell and its transcription, the nucleus also serves as a mechanical gauge of different physical and topographical features of the tissue microenvironment. In this review, we delve into the intricate mechanisms by which the nucleus and different ion channels regulate cell migration in confinement. We review evidence suggesting an interplay between macromolecular nuclear-cytoplasmic transport (NCT) and ionic transport across the cell membrane during confined migration. We also discuss the roles of the nucleus and ion channel-mediated mechanosensation, whether acting independently or in tandem, in orchestrating migratory mechanoresponses. Understanding nuclear and ion channel sensing, and their crosstalk, is critical to advancing our knowledge of cell migration in health and disease.
Collapse
Affiliation(s)
| | - Emily O Wisniewski
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Bishwa R Si
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Petr Kalab
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA.
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, The Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
7
|
Ju RJ, Falconer AD, Schmidt CJ, Enriquez Martinez MA, Dean KM, Fiolka RP, Sester DP, Nobis M, Timpson P, Lomakin AJ, Danuser G, White MD, Haass NK, Oelz DB, Stehbens SJ. Compression-dependent microtubule reinforcement enables cells to navigate confined environments. Nat Cell Biol 2024; 26:1520-1534. [PMID: 39160291 DOI: 10.1038/s41556-024-01476-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/11/2024] [Indexed: 08/21/2024]
Abstract
Cells migrating through complex three-dimensional environments experience considerable physical challenges, including tensile stress and compression. To move, cells need to resist these forces while also squeezing the large nucleus through confined spaces. This requires highly coordinated cortical contractility. Microtubules can both resist compressive forces and sequester key actomyosin regulators to ensure appropriate activation of contractile forces. Yet, how these two roles are integrated to achieve nuclear transmigration in three dimensions is largely unknown. Here, we demonstrate that compression triggers reinforcement of a dedicated microtubule structure at the rear of the nucleus by the mechanoresponsive recruitment of cytoplasmic linker-associated proteins, which dynamically strengthens and repairs the lattice. These reinforced microtubules form the mechanostat: an adaptive feedback mechanism that allows the cell to both withstand compressive force and spatiotemporally organize contractility signalling pathways. The microtubule mechanostat facilitates nuclear positioning and coordinates force production to enable the cell to pass through constrictions. Disruption of the mechanostat imbalances cortical contractility, stalling migration and ultimately resulting in catastrophic cell rupture. Our findings reveal a role for microtubules as cellular sensors that detect and respond to compressive forces, enabling movement and ensuring survival in mechanically demanding environments.
Collapse
Affiliation(s)
- Robert J Ju
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Alistair D Falconer
- School of Mathematics and Physics, University of Queensland, Brisbane, Queensland, Australia
| | - Christanny J Schmidt
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Marco A Enriquez Martinez
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Kevin M Dean
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. and Ida Green Centre for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Reto P Fiolka
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. and Ida Green Centre for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David P Sester
- TRI Flow Cytometry Suite (TRI.fcs), Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Max Nobis
- Faculty of Medicine, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
- Faculty of Medicine, St. Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Paul Timpson
- Faculty of Medicine, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
- Faculty of Medicine, St. Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Alexis J Lomakin
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
- Institute of Medical Chemistry and Pathobiochemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. and Ida Green Centre for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Melanie D White
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Nikolas K Haass
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Dietmar B Oelz
- School of Mathematics and Physics, University of Queensland, Brisbane, Queensland, Australia.
| | - Samantha J Stehbens
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia.
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
8
|
Lee SH, Yousafzai MS, Mohler K, Yadav V, Amiri S, Szuszkiewicz J, Levchenko A, Rinehart J, Murrell M. SPAK-dependent cotransporter activity mediates capillary adhesion and pressure during glioblastoma migration in confined spaces. Mol Biol Cell 2023; 34:ar122. [PMID: 37672340 PMCID: PMC10846615 DOI: 10.1091/mbc.e23-03-0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 09/08/2023] Open
Abstract
The invasive potential of glioblastoma cells is attributed to large changes in pressure and volume, driven by diverse elements, including the cytoskeleton and ion cotransporters. However, how the cell actuates changes in pressure and volume in confinement, and how these changes contribute to invasive motion is unclear. Here, we inhibited SPAK activity, with known impacts on the cytoskeleton and cotransporter activity and explored its role on the migration of glioblastoma cells in confining microchannels to model invasive spread through brain tissue. First, we found that confinement altered cell shape, inducing a transition in morphology that resembled droplet interactions with a capillary vessel, from "wetting" (more adherent) at low confinement, to "nonwetting" (less adherent) at high confinement. This transition was marked by a change from negative to positive pressure by the cells to the confining walls, and an increase in migration speed. Second, we found that the SPAK pathway impacted the migration speed in different ways dependent upon the extent of wetting. For nonwetting cells, SPAK inhibition increased cell-surface tension and cotransporter activity. By contrast, for wetting cells, it also reduced myosin II and YAP phosphorylation. In both cases, membrane-to-cortex attachment is dramatically reduced. Thus, our results suggest that SPAK inhibition differentially coordinates cotransporter and cytoskeleton-induced forces, to impact glioblastoma migration depending on the extent of confinement.
Collapse
Affiliation(s)
- Sung Hoon Lee
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
- Systems Biology Institute, Yale University, West Haven, CT 06516
| | - Muhammad Sulaiman Yousafzai
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
- Systems Biology Institute, Yale University, West Haven, CT 06516
| | - Kyle Mohler
- Systems Biology Institute, Yale University, West Haven, CT 06516
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06510
| | - Vikrant Yadav
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
- Systems Biology Institute, Yale University, West Haven, CT 06516
| | - Sorosh Amiri
- Systems Biology Institute, Yale University, West Haven, CT 06516
- Department of Mechanical Engineering, Yale University, New Haven, CT 06520
| | - Joanna Szuszkiewicz
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
- Systems Biology Institute, Yale University, West Haven, CT 06516
| | - Andre Levchenko
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
- Systems Biology Institute, Yale University, West Haven, CT 06516
| | - Jesse Rinehart
- Systems Biology Institute, Yale University, West Haven, CT 06516
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06510
| | - Michael Murrell
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
- Department of Physics, Yale University, New Haven, CT 06511
- Systems Biology Institute, Yale University, West Haven, CT 06516
| |
Collapse
|
9
|
Zhovmer AS, Manning A, Smith C, Wang J, Ma X, Tsygankov D, Dokholyan NV, Cartagena-Rivera AX, Singh RK, Tabdanov ED. Septins Enable T Cell Contact Guidance via Amoeboid-Mesenchymal Switch. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.559597. [PMID: 37808814 PMCID: PMC10557721 DOI: 10.1101/2023.09.26.559597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Lymphocytes exit circulation and enter in-tissue guided migration toward sites of tissue pathologies, damage, infection, or inflammation. By continuously sensing and adapting to the guiding chemo-mechano-structural properties of the tissues, lymphocytes dynamically alternate and combine their amoeboid (non-adhesive) and mesenchymal (adhesive) migration modes. However, which mechanisms guide and balance different migration modes are largely unclear. Here we report that suppression of septins GTPase activity induces an abrupt amoeboid-to-mesenchymal transition of T cell migration mode, characterized by a distinct, highly deformable integrin-dependent immune cell contact guidance. Surprisingly, the T cell actomyosin cortex contractility becomes diminished, dispensable and antagonistic to mesenchymal-like migration mode. Instead, mesenchymal-like T cells rely on microtubule stabilization and their non-canonical dynein motor activity for high fidelity contact guidance. Our results establish septin's GTPase activity as an important on/off switch for integrin-dependent migration of T lymphocytes, enabling their dynein-driven fluid-like mesenchymal propulsion along the complex adhesion cues.
Collapse
Affiliation(s)
- Alexander S Zhovmer
- Center for Biologics Evaluation & Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Alexis Manning
- Center for Biologics Evaluation & Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Chynna Smith
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Jian Wang
- Departments of Pharmacology, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Xuefei Ma
- Center for Biologics Evaluation & Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Denis Tsygankov
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Nikolay V Dokholyan
- Departments of Pharmacology, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, USA
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, The Pennsylvania State University Hershey-Hummelstown, PA, USA
| | - Alexander X Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Rakesh K Singh
- Department of Obstetrics & Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Erdem D Tabdanov
- Departments of Pharmacology, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, USA
- Penn State Cancer Institute, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| |
Collapse
|
10
|
Shinde P, Kiepas A, Zhang L, Sudhir S, Konstantopoulos K, Stamatos NM. Polysialylation controls immune function of myeloid cells in murine model of pneumococcal pneumonia. Cell Rep 2023; 42:112648. [PMID: 37339052 PMCID: PMC10592499 DOI: 10.1016/j.celrep.2023.112648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/13/2023] [Accepted: 05/31/2023] [Indexed: 06/22/2023] Open
Abstract
Polysialic acid (polySia) is a post-translational modification of a select group of cell-surface proteins that guides cellular interactions. As the overall impact of changes in expression of this glycan on leukocytes during infection is not known, we evaluate the immune response of polySia-deficient ST8SiaIV-/- mice infected with Streptococcus pneumoniae (Spn). Compared with wild-type (WT) mice, ST8SiaIV-/- mice are less susceptible to infection and clear Spn from airways faster, with alveolar macrophages demonstrating greater viability and phagocytic activity. Leukocyte pulmonary recruitment, paradoxically, is diminished in infected ST8SiaIV-/- mice, corroborated by adoptive cell transfer, microfluidic migration experiments, and intravital microscopy, and possibly explained by dysregulated ERK1/2 signaling. PolySia is progressively lost from neutrophils and monocytes migrating from bone marrow to alveoli in Spn-infected WT mice, consistent with changing cellular functions. These data highlight multidimensional effects of polySia on leukocytes during an immune response and suggest therapeutic interventions for optimizing immunity.
Collapse
Affiliation(s)
- Prajakta Shinde
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Alexander Kiepas
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Lei Zhang
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Shreya Sudhir
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Nicholas M Stamatos
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
11
|
A.Alamir HT, Ismaeel GL, Jalil AT, Hadi WH, Jasim IK, Almulla AF, Radhea ZA. Advanced injectable hydrogels for bone tissue regeneration. Biophys Rev 2023; 15:223-237. [PMID: 37124921 PMCID: PMC10133430 DOI: 10.1007/s12551-023-01053-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/17/2023] [Indexed: 05/02/2023] Open
Abstract
Diseases or defects of the skeleton are hazardous because of their specificity and intricacy. Bone tissue engineering has become an important area of research that offers promising new tools for making biomimetic hydrogels that can be used to treat bone diseases. New hydrogels with a distinctive 3D network structure, high water content, and functional capabilities are ranked among the most promising candidates for bone tissue engineering. This makes them helpful in treating cartilage injury, skull deformity, and arthritis. This review will briefly introduce the variety of biocompatible functional hydrogels used in cell culture and bone tissue regeneration. Many gel design concepts, such as crosslinking procedures, controlled release properties, and alternative bionic methodology, were stressed regarding injectable hydrogels to form bone tissue. Hydrogels manufactured from biocompatible materials are a promising option for minimally invasive surgery because of their adaptable physicochemical qualities, ability to fill irregularly shaped defect sites, and ability to grow hormones or release drugs in response to external stimuli. Also included in this overview is a quick rundown of the more practical designs employed in treating bone disorders. Essential details on injectable hydrogel scaffolds for bone tissue regeneration are described in this article.
Collapse
Affiliation(s)
| | | | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Hilla, Babylon, 51001 Iraq
| | | | - Ihsan K. Jasim
- Department of Pharmacology, Al-Turath University College, Baghdad, Iraq
| | - Abbas F. Almulla
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | | |
Collapse
|
12
|
Law RA, Kiepas A, Desta HE, Perez Ipiña E, Parlani M, Lee SJ, Yankaskas CL, Zhao R, Mistriotis P, Wang N, Gu Z, Kalab P, Friedl P, Camley BA, Konstantopoulos K. Cytokinesis machinery promotes cell dissociation from collectively migrating strands in confinement. SCIENCE ADVANCES 2023; 9:eabq6480. [PMID: 36630496 PMCID: PMC9833664 DOI: 10.1126/sciadv.abq6480] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 12/07/2022] [Indexed: 05/10/2023]
Abstract
Cells tune adherens junction dynamics to regulate epithelial integrity in diverse (patho)physiological processes, including cancer metastasis. We hypothesized that the spatially confining architecture of peritumor stroma promotes metastatic cell dissemination by remodeling cell-cell adhesive interactions. By combining microfluidics with live-cell imaging, FLIM/FRET biosensors, and optogenetic tools, we show that confinement induces leader cell dissociation from cohesive ensembles. Cell dissociation is triggered by myosin IIA (MIIA) dismantling of E-cadherin cell-cell junctions, as recapitulated by a mathematical model. Elevated MIIA contractility is controlled by RhoA/ROCK activation, which requires distinct guanine nucleotide exchange factors (GEFs). Confinement activates RhoA via nucleocytoplasmic shuttling of the cytokinesis-regulatory proteins RacGAP1 and Ect2 and increased microtubule dynamics, which results in the release of active GEF-H1. Thus, confining microenvironments are sufficient to induce cell dissemination from primary tumors by remodeling E-cadherin cell junctions via the interplay of microtubules, nuclear trafficking, and RhoA/ROCK/MIIA pathway and not by down-regulating E-cadherin expression.
Collapse
Affiliation(s)
- Robert A. Law
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Alexander Kiepas
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Habben E. Desta
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Emiliano Perez Ipiña
- William H. Miller III Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Maria Parlani
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Se Jong Lee
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Christopher L. Yankaskas
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Runchen Zhao
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Panagiotis Mistriotis
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Nianchao Wang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Zhizhan Gu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Petr Kalab
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Peter Friedl
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Cancer Genomics Center, 3584 Utrecht, Netherlands
| | - Brian A. Camley
- William H. Miller III Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
13
|
Yu Y, Yu T, Wang X, Liu D. Functional Hydrogels and Their Applications in Craniomaxillofacial Bone Regeneration. Pharmaceutics 2022; 15:pharmaceutics15010150. [PMID: 36678779 PMCID: PMC9864650 DOI: 10.3390/pharmaceutics15010150] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Craniomaxillofacial bone defects are characterized by an irregular shape, bacterial and inflammatory environment, aesthetic requirements, and the need for the functional recovery of oral-maxillofacial areas. Conventional clinical treatments are currently unable to achieve high-quality craniomaxillofacial bone regeneration. Hydrogels are a class of multifunctional platforms made of polymers cross-linked with high water content, good biocompatibility, and adjustable physicochemical properties for the intelligent delivery of goods. These characteristics make hydrogel systems a bright prospect for clinical applications in craniomaxillofacial bone. In this review, we briefly demonstrate the properties of hydrogel systems that can come into effect in the field of bone regeneration. In addition, we summarize the hydrogel systems that have been developed for craniomaxillofacial bone regeneration in recent years. Finally, we also discuss the prospects in the field of craniomaxillofacial bone tissue engineering; these discussions can serve as an inspiration for future hydrogel design.
Collapse
Affiliation(s)
- Yi Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Tingting Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- Correspondence: (X.W.); (D.L.)
| | - Dawei Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China
- National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
- Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
- Correspondence: (X.W.); (D.L.)
| |
Collapse
|
14
|
Hu S, Liu T, Xue C, Li Y, Yang Y, Xu X, Liu B, Chen X, Zhao Y, Qin K. A high-throughput microfluidic device inspired by the Wheatstone bridge principle for characterizing the mechanical properties of single cells. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2022; 14:4813-4821. [PMID: 36382629 DOI: 10.1039/d2ay01416e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The mechanical properties of single cells have been recognized as biomarkers for identifying individual cells and diagnosing human diseases. Microfluidic devices based on the flow cytometry principle, which are not limited by the vision field of a microscope and can achieve a very high throughput, have been extensively adopted to measure the mechanical properties of single cells. However, these kinds of microfluidic devices usually required pressure-driven pumps with a very low flow rate and high precision. In this study, we developed a high-throughput microfluidic device inspired by the Wheatstone bridge principle for characterizing the mechanical properties of single cells. The microfluidic analogue of the Wheatstone bridge not only took advantage of flow cytometry, but also allowed precise control of a very low flow rate through the constricted channel with a higher input flow rate generated by a commercially available pressure-driven pump. Under different input flow rates of the pump, the apparent elastic moduli and the fluidity of osteosarcoma (U-2OS) cells and cervical carcinoma (HeLa) cells were measured by monitoring their dynamic deformations passing through the bridge-channel with different sizes of rectangular constrictions. The results showed that the input flow rate had little effect on measuring the mechanical properties of the cells, while the ratio of cell radius to effective constriction radius was different, i.e., for U-2OS cells it was 1.20 and for HeLa cells it was 1.09. Under this condition compared with predecessors, our statistic results of cell mechanical properties exhibited minimal errors. Furthermore, the cell viability after measurements was kept above 90% that demonstrated the non-destructive property of our proposed method.
Collapse
Affiliation(s)
- Siyu Hu
- School of Mechanical Engineering, Dalian University of Technology, Dalian 116024, Liaoning Province, China
| | - Tianmian Liu
- DUT-BSU Joint Institute, Dalian University of Technology, Dalian 116024, Liaoning Province, China
| | - Chundong Xue
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| | - Yongjiang Li
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| | - Yunong Yang
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian 116024, Liaoning Province, China
| | - Xing Xu
- Department of Endoscopy, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning Province, China
| | - Bo Liu
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian 116024, Liaoning Province, China
| | - Xiaoming Chen
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| | - Yan Zhao
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian 116024, Liaoning Province, China
- Department of Gastric Cancer, Liaoning Cancer Hospital and Institute, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, Liaoning Province, China
| | - Kairong Qin
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian 116024, Liaoning Province, China
| |
Collapse
|
15
|
Pillai S, Munguia-Lopez JG, Tran SD. Hydrogels for Salivary Gland Tissue Engineering. Gels 2022; 8:730. [PMID: 36354638 PMCID: PMC9690182 DOI: 10.3390/gels8110730] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/28/2022] [Accepted: 11/07/2022] [Indexed: 09/19/2023] Open
Abstract
Mimicking the complex architecture of salivary glands (SGs) outside their native niche is challenging due their multicellular and highly branched organization. However, significant progress has been made to recapitulate the gland structure and function using several in vitro and ex vivo models. Hydrogels are polymers with the potential to retain a large volume of water inside their three-dimensional structure, thus simulating extracellular matrix properties that are essential for the cell and tissue integrity. Hydrogel-based culture of SG cells has seen a tremendous success in terms of developing platforms for cell expansion, building an artificial gland, and for use in transplantation to rescue loss of SG function. Both natural and synthetic hydrogels have been used widely in SG tissue engineering applications owing to their properties that support the proliferation, reorganization, and polarization of SG epithelial cells. While recent improvements in hydrogel properties are essential to establish more sophisticated models, the emphasis should still be made towards supporting factors such as mechanotransduction and associated signaling cues. In this concise review, we discuss considerations of an ideal hydrogel-based biomaterial for SG engineering and their associated signaling pathways. We also discuss the current advances made in natural and synthetic hydrogels for SG tissue engineering applications.
Collapse
Affiliation(s)
| | | | - Simon D. Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, 3640 Rue University, Montreal, QC H3A 0C7, Canada
| |
Collapse
|
16
|
Bahr JC, Li XY, Feinberg TY, Jiang L, Weiss SJ. Divergent regulation of basement membrane trafficking by human macrophages and cancer cells. Nat Commun 2022; 13:6409. [PMID: 36302921 PMCID: PMC9613642 DOI: 10.1038/s41467-022-34087-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/13/2022] [Indexed: 12/25/2022] Open
Abstract
Macrophages and cancer cells populations are posited to navigate basement membrane barriers by either mobilizing proteolytic enzymes or deploying mechanical forces. Nevertheless, the relative roles, or identity, of the proteinase -dependent or -independent mechanisms used by macrophages versus cancer cells to transmigrate basement membrane barriers harboring physiologically-relevant covalent crosslinks remains ill-defined. Herein, both macrophages and cancer cells are shown to mobilize membrane-anchored matrix metalloproteinases to proteolytically remodel native basement membranes isolated from murine tissues while infiltrating the underlying interstitial matrix ex vivo. In the absence of proteolytic activity, however, only macrophages deploy actomyosin-generated forces to transmigrate basement membrane pores, thereby providing the cells with proteinase-independent access to the interstitial matrix while simultaneously exerting global effects on the macrophage transcriptome. By contrast, cancer cell invasive activity is reliant on metalloproteinase activity and neither mechanical force nor changes in nuclear rigidity rescue basement membrane transmigration. These studies identify membrane-anchored matrix metalloproteinases as key proteolytic effectors of basement membrane remodeling by macrophages and cancer cells while also defining the divergent invasive strategies used by normal and neoplastic cells to traverse native tissue barriers.
Collapse
Affiliation(s)
- Julian C Bahr
- Cancer Biology Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xiao-Yan Li
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tamar Y Feinberg
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Long Jiang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stephen J Weiss
- Cancer Biology Graduate Program, University of Michigan, Ann Arbor, MI, 48109, USA.
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
- Division of Genetic Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
17
|
Zhang Y, Li Y, Thompson KN, Stoletov K, Yuan Q, Bera K, Lee SJ, Zhao R, Kiepas A, Wang Y, Mistriotis P, Serra SA, Lewis JD, Valverde MA, Martin SS, Sun SX, Konstantopoulos K. Polarized NHE1 and SWELL1 regulate migration direction, efficiency and metastasis. Nat Commun 2022; 13:6128. [PMID: 36253369 PMCID: PMC9576788 DOI: 10.1038/s41467-022-33683-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 09/26/2022] [Indexed: 12/24/2022] Open
Abstract
Cell migration regulates diverse (patho)physiological processes, including cancer metastasis. According to the Osmotic Engine Model, polarization of NHE1 at the leading edge of confined cells facilitates water uptake, cell protrusion and motility. The physiological relevance of the Osmotic Engine Model and the identity of molecules mediating cell rear shrinkage remain elusive. Here, we demonstrate that NHE1 and SWELL1 preferentially polarize at the cell leading and trailing edges, respectively, mediate cell volume regulation, cell dissemination from spheroids and confined migration. SWELL1 polarization confers migration direction and efficiency, as predicted mathematically and determined experimentally via optogenetic spatiotemporal regulation. Optogenetic RhoA activation at the cell front triggers SWELL1 re-distribution and migration direction reversal in SWELL1-expressing, but not SWELL1-knockdown, cells. Efficient cell reversal also requires Cdc42, which controls NHE1 repolarization. Dual NHE1/SWELL1 knockdown inhibits breast cancer cell extravasation and metastasis in vivo, thereby illustrating the physiological significance of the Osmotic Engine Model.
Collapse
Grants
- R01 CA254193 NCI NIH HHS
- R01 GM134542 NIGMS NIH HHS
- This work was supported, in part, by an NIH/NCI R01 CA254193 (K.K., S.S.M., S.X.S.), R01 GM134542 (S.X.S., K.K.), NSF 2045715 (Y.L.), the Spanish Ministry of Science, Education and Universities through grants RTI2018-099718-B-100 (M.A.V.), an institutional “Maria de Maeztu” Programme for Units of Excellence in R&D and FEDER funds (M.A.V.) and postdoctoral fellowships from the Fonds de recherche du Quebec - Nature et technologies and the Natural Sciences and Engineering Research Council of Canada (A.K.). The opinions, findings, and conclusions, or recommendations expressed are those of the authors and do not necessarily reflect the views of any of the funding agencies.
Collapse
Affiliation(s)
- Yuqi Zhang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Yizeng Li
- Department of Biomedical Engineering, Binghamton University, SUNY, Binghamton, NY, 13902, USA
| | - Keyata N Thompson
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Konstantin Stoletov
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Qinling Yuan
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Kaustav Bera
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Se Jong Lee
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Runchen Zhao
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Alexander Kiepas
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Yao Wang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Panagiotis Mistriotis
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Chemical Engineering, Auburn University, Auburn, AL, 36849, USA
| | - Selma A Serra
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain
| | - John D Lewis
- Department of Oncology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Miguel A Valverde
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain
| | - Stuart S Martin
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Sean X Sun
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA.
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA.
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA.
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Oncology, The Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
18
|
Ma Y, Wang X, Su T, Lu F, Chang Q, Gao J. Recent Advances in Macroporous Hydrogels for Cell Behavior and Tissue Engineering. Gels 2022; 8:606. [PMID: 36286107 PMCID: PMC9601978 DOI: 10.3390/gels8100606] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/07/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022] Open
Abstract
Hydrogels have been extensively used as scaffolds in tissue engineering for cell adhesion, proliferation, migration, and differentiation because of their high-water content and biocompatibility similarity to the extracellular matrix. However, submicron or nanosized pore networks within hydrogels severely limit cell survival and tissue regeneration. In recent years, the application of macroporous hydrogels in tissue engineering has received considerable attention. The macroporous structure not only facilitates nutrient transportation and metabolite discharge but also provides more space for cell behavior and tissue formation. Several strategies for creating and functionalizing macroporous hydrogels have been reported. This review began with an overview of the advantages and challenges of macroporous hydrogels in the regulation of cellular behavior. In addition, advanced methods for the preparation of macroporous hydrogels to modulate cellular behavior were discussed. Finally, future research in related fields was discussed.
Collapse
Affiliation(s)
| | | | | | | | - Qiang Chang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou 510515, China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou 510515, China
| |
Collapse
|
19
|
Bera K, Kiepas A, Zhang Y, Sun SX, Konstantopoulos K. The interplay between physical cues and mechanosensitive ion channels in cancer metastasis. Front Cell Dev Biol 2022; 10:954099. [PMID: 36158191 PMCID: PMC9490090 DOI: 10.3389/fcell.2022.954099] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Physical cues have emerged as critical influencers of cell function during physiological processes, like development and organogenesis, and throughout pathological abnormalities, including cancer progression and fibrosis. While ion channels have been implicated in maintaining cellular homeostasis, their cell surface localization often places them among the first few molecules to sense external cues. Mechanosensitive ion channels (MICs) are especially important transducers of physical stimuli into biochemical signals. In this review, we describe how physical cues in the tumor microenvironment are sensed by MICs and contribute to cancer metastasis. First, we highlight mechanical perturbations, by both solid and fluid surroundings typically found in the tumor microenvironment and during critical stages of cancer cell dissemination from the primary tumor. Next, we describe how Piezo1/2 and transient receptor potential (TRP) channels respond to these physical cues to regulate cancer cell behavior during different stages of metastasis. We conclude by proposing alternative mechanisms of MIC activation that work in tandem with cytoskeletal components and other ion channels to bestow cells with the capacity to sense, respond and navigate through the surrounding microenvironment. Collectively, this review provides a perspective for devising treatment strategies against cancer by targeting MICs that sense aberrant physical characteristics during metastasis, the most lethal aspect of cancer.
Collapse
Affiliation(s)
- Kaustav Bera
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
| | - Alexander Kiepas
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Alexander Kiepas, ; Konstantinos Konstantopoulos,
| | - Yuqi Zhang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
| | - Sean X. Sun
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, The Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Alexander Kiepas, ; Konstantinos Konstantopoulos,
| |
Collapse
|
20
|
Jana A, Tran A, Gill A, Kiepas A, Kapania RK, Konstantopoulos K, Nain AS. Sculpting Rupture-Free Nuclear Shapes in Fibrous Environments. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203011. [PMID: 35863910 PMCID: PMC9443471 DOI: 10.1002/advs.202203011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Indexed: 05/07/2023]
Abstract
Cytoskeleton-mediated force transmission regulates nucleus morphology. How nuclei shaping occurs in fibrous in vivo environments remains poorly understood. Here suspended nanofiber networks of precisely tunable (nm-µm) diameters are used to quantify nucleus plasticity in fibrous environments mimicking the natural extracellular matrix. Contrary to the apical cap over the nucleus in cells on 2-dimensional surfaces, the cytoskeleton of cells on fibers displays a uniform actin network caging the nucleus. The role of contractility-driven caging in sculpting nuclear shapes is investigated as cells spread on aligned single fibers, doublets, and multiple fibers of varying diameters. Cell contractility increases with fiber diameter due to increased focal adhesion clustering and density of actin stress fibers, which correlates with increased mechanosensitive transcription factor Yes-associated protein (YAP) translocation to the nucleus. Unexpectedly, large- and small-diameter fiber combinations lead to teardrop-shaped nuclei due to stress fiber anisotropy across the cell. As cells spread on fibers, diameter-dependent nuclear envelope invaginations that run the nucleus's length are formed at fiber contact sites. The sharpest invaginations enriched with heterochromatin clustering and sites of DNA repair are insufficient to trigger nucleus rupture. Overall, the authors quantitate the previously unknown sculpting and adaptability of nuclei to fibrous environments with pathophysiological implications.
Collapse
Affiliation(s)
- Aniket Jana
- Department of Mechanical EngineeringVirginia TechBlacksburgVA24061USA
| | - Avery Tran
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Amritpal Gill
- Department of Mechanical EngineeringVirginia TechBlacksburgVA24061USA
| | - Alexander Kiepas
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Rakesh K. Kapania
- Kevin T. Crofton Department of Aerospace EngineeringVirginia TechBlacksburgVA24061USA
| | | | - Amrinder S. Nain
- Department of Mechanical EngineeringVirginia TechBlacksburgVA24061USA
| |
Collapse
|
21
|
Lee SH, Hou JC, Hamidzadeh A, Yousafzai MS, Ajeti V, Chang H, Odde DJ, Murrell M, Levchenko A. A molecular clock controls periodically driven cell migration in confined spaces. Cell Syst 2022; 13:514-529.e10. [PMID: 35679858 DOI: 10.1016/j.cels.2022.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/10/2021] [Accepted: 05/13/2022] [Indexed: 01/25/2023]
Abstract
Navigation through a dense, physically confining extracellular matrix is common in invasive cell spread and tissue reorganization but is still poorly understood. Here, we show that this migration is mediated by cyclic changes in the activity of a small GTPase RhoA, which is dependent on the oscillatory changes in the activity and abundance of the RhoA guanine nucleotide exchange factor, GEF-H1, and triggered by a persistent increase in the intracellular Ca2+ levels. We show that the molecular clock driving these cyclic changes is mediated by two coupled negative feedback loops, dependent on the microtubule dynamics, with a frequency that can be experimentally modulated based on a predictive mathematical model. We further demonstrate that an increasing frequency of the clock translates into a faster cell migration within physically confining spaces. This work lays the foundation for a better understanding of the molecular mechanisms dynamically driving cell migration in complex environments.
Collapse
Affiliation(s)
- Sung Hoon Lee
- Yale Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Jay C Hou
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Archer Hamidzadeh
- Yale Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - M Sulaiman Yousafzai
- Yale Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA; Department of Physics, Yale University, New Haven, CT 06520, USA
| | - Visar Ajeti
- Yale Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA; Department of Physics, Yale University, New Haven, CT 06520, USA; Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Hao Chang
- Yale Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael Murrell
- Yale Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA; Department of Physics, Yale University, New Haven, CT 06520, USA
| | - Andre Levchenko
- Yale Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
22
|
Moriarty RA, Mili S, Stroka KM. RNA localization in confined cells depends on cellular mechanical activity and contributes to confined migration. iScience 2022; 25:103845. [PMID: 35198898 PMCID: PMC8850802 DOI: 10.1016/j.isci.2022.103845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/30/2021] [Accepted: 01/27/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer cells experience mechanical confining forces during metastasis and, consequently, can alter their migratory mechanisms. Localization of numerous mRNAs to cell protrusions contributes to cell polarization and migration and is controlled by proteins that can bind RNA and/or cytoskeletal elements, such as the adenomatous polyposis coli (APC). Here, we demonstrate that peripheral localization of APC-dependent RNAs in cells within confined microchannels is cell type dependent. This varying phenotype is determined by the levels of a detyrosinated tubulin network. We show that this network is regulated by mechanoactivity and that cells with mechanosensitive ion channels and increased myosin II activity direct peripheral localization of the RAB13 APC-dependent RNA. Through specific mislocalization of the RAB13 RNA, we show that peripheral RNA localization contributes to confined cell migration. Our results indicate that a cell’s mechanical activity determines its ability to peripherally target RNAs and utilize them for movement in confinement. Peripheral localization of APC-dependent RNAs in confinement depends on cell type RNA localization in confined cells is controlled by the mechanoactivity of cells RNA localization phenotype is influenced by the detyrosinated tubulin network Peripheral RNA accumulation functionally contributes to confined cell migration
Collapse
Affiliation(s)
- Rebecca A. Moriarty
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Fischell Department of Bioengineering, University of Maryland College Park, College Park, MD 20742, USA
| | - Stavroula Mili
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Corresponding author
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland College Park, College Park, MD 20742, USA
- Maryland Biophysics Program, University of Maryland College Park, College Park, MD 20742, USA
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland Baltimore, Baltimore, MD 21202, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore, Baltimore, MD 21202, USA
- Corresponding author
| |
Collapse
|
23
|
Tuntithavornwat S, Shea DJ, Wong BS, Guardia T, Lee SJ, Yankaskas CL, Zheng L, Kontrogianni-Konstantopoulos A, Konstantopoulos K. Giant obscurin regulates migration and metastasis via RhoA-dependent cytoskeletal remodeling in pancreatic cancer. Cancer Lett 2022; 526:155-167. [PMID: 34826548 PMCID: PMC9427004 DOI: 10.1016/j.canlet.2021.11.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/15/2021] [Accepted: 11/12/2021] [Indexed: 12/23/2022]
Abstract
Obscurins, encoded by the OBSCN gene, are giant cytoskeletal proteins with structural and regulatory roles. Large scale omics analyses reveal that OBSCN is highly mutated across different types of cancer, exhibiting a 5-8% mutation frequency in pancreatic cancer. Yet, the functional role of OBSCN in pancreatic cancer progression and metastasis has to be delineated. We herein show that giant obscurins are highly expressed in normal pancreatic tissues, but their levels are markedly reduced in pancreatic ductal adenocarcinomas. Silencing of giant obscurins in non-tumorigenic Human Pancreatic Ductal Epithelial (HPDE) cells and obscurin-expressing Panc5.04 pancreatic cancer cells induces an elongated, spindle-like morphology and faster cell migration via cytoskeletal remodeling. Specifically, depletion of giant obscurins downregulates RhoA activity, which in turn results in reduced focal adhesion density, increased microtubule growth rate and faster actin dynamics. Although OBSCN knockdown is not sufficient to induce de novo tumorigenesis, it potentiates tumor growth in a subcutaneous implantation model and exacerbates metastasis in a hemispleen murine model of pancreatic cancer metastasis, thereby shortening survival. Collectively, these findings reveal a critical role of giant obscurins as tumor suppressors in normal pancreatic epithelium whose loss of function induces RhoA-dependent cytoskeletal remodeling, and promotes cell migration, tumor growth and metastasis.
Collapse
Affiliation(s)
- Soontorn Tuntithavornwat
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Daniel J Shea
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Bin Sheng Wong
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA; Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, USA
| | - Talia Guardia
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Se Jong Lee
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Christopher L Yankaskas
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA; Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, USA
| | - Lei Zheng
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aikaterini Kontrogianni-Konstantopoulos
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, USA; Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
24
|
Luo M, Cai G, Ho KKY, Wen K, Tong Z, Deng L, Liu AP. Compression enhances invasive phenotype and matrix degradation of breast Cancer cells via Piezo1 activation. BMC Mol Cell Biol 2022; 23:1. [PMID: 34979904 PMCID: PMC8722159 DOI: 10.1186/s12860-021-00401-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022] Open
Abstract
Background Uncontrolled growth in solid breast cancer generates mechanical compression that may drive the cancer cells into a more invasive phenotype, but little is known about how such compression affects the key events and corresponding regulatory mechanisms associated with invasion of breast cancer cells including cellular behaviors and matrix degradation. Results Here we show that compression enhanced invasion and matrix degradation of breast cancer cells. We also identified Piezo1 as the putative mechanosensitive cellular component that transmitted compression to not only enhance the invasive phenotype, but also induce calcium influx and downstream Src signaling. Furthermore, we demonstrated that Piezo1 was mainly localized in caveolae, and both Piezo1 expression and compression-enhanced invasive phenotype of the breast cancer cells were reduced when caveolar integrity was compromised by either knocking down caveolin1 expression or depleting cholesterol content. Conclusions Taken together, our data indicate that mechanical compression activates Piezo1 channels to mediate enhanced breast cancer cell invasion, which involves both cellular events and matrix degradation. This may be a critical mechanotransduction pathway during breast cancer metastasis, and thus potentially a novel therapeutic target for the disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-021-00401-6.
Collapse
Affiliation(s)
- Mingzhi Luo
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, People's Republic of China.,Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Grace Cai
- Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| | - Kenneth K Y Ho
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.,Present address: Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Kang Wen
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, People's Republic of China
| | - Zhaowen Tong
- Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| | - Linhong Deng
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, Jiangsu, People's Republic of China.
| | - Allen P Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA. .,Applied Physics Program, University of Michigan, Ann Arbor, MI, USA. .,Department of Biophysics, University of Michigan, Ann Arbor, MI, USA. .,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA. .,Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
Cao H, Duan L, Zhang Y, Cao J, Zhang K. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct Target Ther 2021; 6:426. [PMID: 34916490 PMCID: PMC8674418 DOI: 10.1038/s41392-021-00830-x] [Citation(s) in RCA: 410] [Impact Index Per Article: 102.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/05/2023] Open
Abstract
Hydrogel is a type of versatile platform with various biomedical applications after rational structure and functional design that leverages on material engineering to modulate its physicochemical properties (e.g., stiffness, pore size, viscoelasticity, microarchitecture, degradability, ligand presentation, stimulus-responsive properties, etc.) and influence cell signaling cascades and fate. In the past few decades, a plethora of pioneering studies have been implemented to explore the cell-hydrogel matrix interactions and figure out the underlying mechanisms, paving the way to the lab-to-clinic translation of hydrogel-based therapies. In this review, we first introduced the physicochemical properties of hydrogels and their fabrication approaches concisely. Subsequently, the comprehensive description and deep discussion were elucidated, wherein the influences of different hydrogels properties on cell behaviors and cellular signaling events were highlighted. These behaviors or events included integrin clustering, focal adhesion (FA) complex accumulation and activation, cytoskeleton rearrangement, protein cyto-nuclei shuttling and activation (e.g., Yes-associated protein (YAP), catenin, etc.), cellular compartment reorganization, gene expression, and further cell biology modulation (e.g., spreading, migration, proliferation, lineage commitment, etc.). Based on them, current in vitro and in vivo hydrogel applications that mainly covered diseases models, various cell delivery protocols for tissue regeneration and disease therapy, smart drug carrier, bioimaging, biosensor, and conductive wearable/implantable biodevices, etc. were further summarized and discussed. More significantly, the clinical translation potential and trials of hydrogels were presented, accompanied with which the remaining challenges and future perspectives in this field were emphasized. Collectively, the comprehensive and deep insights in this review will shed light on the design principles of new biomedical hydrogels to understand and modulate cellular processes, which are available for providing significant indications for future hydrogel design and serving for a broad range of biomedical applications.
Collapse
Affiliation(s)
- Huan Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Lixia Duan
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Yan Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Jun Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China.
| | - Kun Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China.
| |
Collapse
|
26
|
Gabbireddy SR, Vosatka KW, Chung AJ, Logue JS. Melanoma cells adopt features of both mesenchymal and amoeboid migration within confining channels. Sci Rep 2021; 11:17804. [PMID: 34493759 PMCID: PMC8423822 DOI: 10.1038/s41598-021-97348-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 08/18/2021] [Indexed: 11/10/2022] Open
Abstract
For metastasis to occur, cancer cells must traverse a range of tissue environments. In part, this is accomplished by cells adjusting their migration mode to one that is best suited to the environment. Melanoma cells have been shown to be particularly plastic, frequently using both mesenchymal and amoeboid (bleb-based) modes of migration. It has been demonstrated that 2D confinement will promote the transition from mesenchymal to bleb-based migration. However, if melanoma cells similarly transition to bleb-based migration in response to 3D confinement, such as within narrow channels, is unknown. Here, using micro-fabricated channels, we demonstrate that metastatic, A375-M2, melanoma cells adopt features of both mesenchymal and bleb-based migration. In narrow (8 µm; height and width) channels coated with fibronectin, ~ 50% of melanoma cells were found to use either mesenchymal or bleb-based migration modes. In contrast, the inhibition of Src family kinases or coating channels with BSA, completely eliminated any features of mesenchymal migration. Detailed comparisons of migration parameters revealed that blebbing cells, particularly in the absence of adhesions, were faster than mesenchymal cells. In contrast to what has been previously shown under conditions of 2D confinement, pharmacologically inhibiting Arp2/3 promoted a fast filopodial-based mode of migration. Accordingly, we report that melanoma cells adopt a unique range of phenotypes under conditions of 3D confinement.
Collapse
Affiliation(s)
- Sairisheel R Gabbireddy
- Undergraduate Research Program, Rensselaer Polytechnic Institute (RPI), 110 8th St, Troy, NY, 12180, USA
| | - Karl W Vosatka
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Ave, Albany, NY, 12208, USA
| | - Aram J Chung
- Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute (RPI), 110 8th Street, Troy, NY, 12180, USA
- School of Biomedical Engineering, Korea University, 02841, Seoul, Republic of Korea
- Interdisciplinary Program in Precision Public Health, Korea University, 02841, Seoul, Republic of Korea
| | - Jeremy S Logue
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Ave, Albany, NY, 12208, USA.
| |
Collapse
|
27
|
Yankaskas CL, Bera K, Stoletov K, Serra SA, Carrillo-Garcia J, Tuntithavornwat S, Mistriotis P, Lewis JD, Valverde MA, Konstantopoulos K. The fluid shear stress sensor TRPM7 regulates tumor cell intravasation. SCIENCE ADVANCES 2021; 7:7/28/eabh3457. [PMID: 34244134 PMCID: PMC8270498 DOI: 10.1126/sciadv.abh3457] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/28/2021] [Indexed: 05/09/2023]
Abstract
Tumor cell intravasation preferentially occurs in regions of low fluid shear because high shear is detrimental to tumor cells. Here, we describe a molecular mechanism by which cells avoid high shear during intravasation. The transition from migration to intravasation was modeled using a microfluidic device where cells migrating inside longitudinal tissue-like microchannels encounter an orthogonal channel in which fluid flow induces physiological shear stresses. This approach was complemented with intravital microscopy, patch-clamp, and signal transduction imaging techniques. Fluid shear-induced activation of the transient receptor potential melastatin 7 (TRPM7) channel promotes extracellular calcium influx, which then activates RhoA/myosin-II and calmodulin/IQGAP1/Cdc42 pathways to coordinate reversal of migration direction, thereby avoiding shear stress. Cells displaying higher shear sensitivity due to higher TRPM7 activity levels intravasate less efficiently and establish less invasive metastatic lesions. This study provides a mechanistic interpretation for the role of shear stress and its sensor, TRPM7, in tumor cell intravasation.
Collapse
Affiliation(s)
- Christopher L Yankaskas
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kaustav Bera
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Selma A Serra
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Julia Carrillo-Garcia
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Soontorn Tuntithavornwat
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Panagiotis Mistriotis
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - John D Lewis
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Miguel A Valverde
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA.
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
28
|
Dey K, Roca E, Ramorino G, Sartore L. Progress in the mechanical modulation of cell functions in tissue engineering. Biomater Sci 2021; 8:7033-7081. [PMID: 33150878 DOI: 10.1039/d0bm01255f] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In mammals, mechanics at multiple stages-nucleus to cell to ECM-underlie multiple physiological and pathological functions from its development to reproduction to death. Under this inspiration, substantial research has established the role of multiple aspects of mechanics in regulating fundamental cellular processes, including spreading, migration, growth, proliferation, and differentiation. However, our understanding of how these mechanical mechanisms are orchestrated or tuned at different stages to maintain or restore the healthy environment at the tissue or organ level remains largely a mystery. Over the past few decades, research in the mechanical manipulation of the surrounding environment-known as substrate or matrix or scaffold on which, or within which, cells are seeded-has been exceptionally enriched in the field of tissue engineering and regenerative medicine. To do so, traditional tissue engineering aims at recapitulating key mechanical milestones of native ECM into a substrate for guiding the cell fate and functions towards specific tissue regeneration. Despite tremendous progress, a big puzzle that remains is how the cells compute a host of mechanical cues, such as stiffness (elasticity), viscoelasticity, plasticity, non-linear elasticity, anisotropy, mechanical forces, and mechanical memory, into many biological functions in a cooperative, controlled, and safe manner. High throughput understanding of key cellular decisions as well as associated mechanosensitive downstream signaling pathway(s) for executing these decisions in response to mechanical cues, solo or combined, is essential to address this issue. While many reports have been made towards the progress and understanding of mechanical cues-particularly, substrate bulk stiffness and viscoelasticity-in regulating the cellular responses, a complete picture of mechanical cues is lacking. This review highlights a comprehensive view on the mechanical cues that are linked to modulate many cellular functions and consequent tissue functionality. For a very basic understanding, a brief discussion of the key mechanical players of ECM and the principle of mechanotransduction process is outlined. In addition, this review gathers together the most important data on the stiffness of various cells and ECM components as well as various tissues/organs and proposes an associated link from the mechanical perspective that is not yet reported. Finally, beyond addressing the challenges involved in tuning the interplaying mechanical cues in an independent manner, emerging advances in designing biomaterials for tissue engineering are also explored.
Collapse
Affiliation(s)
- Kamol Dey
- Department of Applied Chemistry and Chemical Engineering, Faculty of Science, University of Chittagong, Bangladesh
| | | | | | | |
Collapse
|
29
|
Aoun L, Nègre P, Gonsales C, Seveau de Noray V, Brustlein S, Biarnes-Pelicot M, Valignat MP, Theodoly O. Leukocyte transmigration and longitudinal forward-thrusting force in a microfluidic Transwell device. Biophys J 2021; 120:2205-2221. [PMID: 33838136 DOI: 10.1016/j.bpj.2021.03.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/10/2021] [Accepted: 03/24/2021] [Indexed: 01/21/2023] Open
Abstract
Transmigration of leukocytes across blood vessels walls is a critical step of the immune response. Transwell assays examine transmigration properties in vitro by counting cells passages through a membrane; however, the difficulty of in situ imaging hampers a clear disentanglement of the roles of adhesion, chemokinesis, and chemotaxis. We used here microfluidic Transwells to image the cells' transition from 2D migration on a surface to 3D migration in a confining microchannel and measure cells longitudinal forward-thrusting force in microchannels. Primary human effector T lymphocytes adhering with integrins LFA-1 (αLβ2) had a marked propensity to transmigrate in Transwells without chemotactic cue. Both adhesion and contractility were important to overcome the critical step of nucleus penetration but were remarkably dispensable for 3D migration in smooth microchannels deprived of topographic features. Transmigration in smooth channels was qualitatively consistent with a propulsion by treadmilling of cell envelope and squeezing of cell trailing edge. Stalling conditions of 3D migration were then assessed by imposing pressure drops across microchannels. Without specific adhesion, the cells slid backward with subnanonewton forces, showing that 3D migration under stress is strongly limited by a lack of adhesion and friction with channels. With specific LFA-1 mediated adhesion, stalling occurred at around 3 and 6 nN in 2 × 4 and 4 × 4 μm2 channels, respectively, supporting that stalling of adherent cells was under pressure control rather than force control. The stall pressure of 4 mbar is consistent with the pressure of actin filament polymerization that mediates lamellipod growth. The arrest of adherent cells under stress therefore seems controlled by the compression of the cell leading edge, which perturbs cells front-rear polarization and triggers adhesion failure or polarization reversal. Although stalling assays in microfluidic Transwells do not mimic in vivo transmigration, they provide a powerful tool to scrutinize 2D and 3D migration, barotaxis, and chemotaxis.
Collapse
Affiliation(s)
- Laurene Aoun
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | - Paulin Nègre
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | - Cristina Gonsales
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | | | - Sophie Brustlein
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | | | - Marie-Pierre Valignat
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France
| | - Olivier Theodoly
- LAI, Aix-Marseille Univ, CNRS, INSERM, Turing Centre for Living Systems, Marseille, France.
| |
Collapse
|
30
|
Zhao R, Cui S, Ge Z, Zhang Y, Bera K, Zhu L, Sun SX, Konstantopoulos K. Hydraulic resistance induces cell phenotypic transition in confinement. SCIENCE ADVANCES 2021; 7:7/17/eabg4934. [PMID: 33893091 PMCID: PMC8064631 DOI: 10.1126/sciadv.abg4934] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/12/2021] [Indexed: 05/24/2023]
Abstract
Cells penetrating into confinement undergo mesenchymal-to-amoeboid transition. The topographical features of the microenvironment expose cells to different hydraulic resistance levels. How cells respond to hydraulic resistance is unknown. We show that the cell phenotype shifts from amoeboid to mesenchymal upon increasing resistance. By combining automated morphological tracking and wavelet analysis along with fluorescence recovery after photobleaching (FRAP), we found an oscillatory phenotypic transition that cycles from blebbing to short, medium, and long actin network formation, and back to blebbing. Elevated hydraulic resistance promotes focal adhesion maturation and long actin filaments, thereby reducing the period required for amoeboid-to-mesenchymal transition. The period becomes independent of resistance upon blocking the mechanosensor TRPM7. Mathematical modeling links intracellular calcium oscillations with actomyosin turnover and force generation and recapitulates experimental data. We identify hydraulic resistance as a critical physical cue controlling cell phenotype and present an approach for connecting fluorescent signal fluctuations to morphological oscillations.
Collapse
Affiliation(s)
- Runchen Zhao
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Siqi Cui
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Zhuoxu Ge
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yuqi Zhang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kaustav Bera
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Lily Zhu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sean X Sun
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
31
|
Puleri DF, Balogh P, Randles A. Computational models of cancer cell transport through the microcirculation. Biomech Model Mechanobiol 2021; 20:1209-1230. [PMID: 33765196 DOI: 10.1007/s10237-021-01452-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/08/2021] [Indexed: 02/07/2023]
Abstract
The transport of cancerous cells through the microcirculation during metastatic spread encompasses several interdependent steps that are not fully understood. Computational models which resolve the cellular-scale dynamics of complex microcirculatory flows offer considerable potential to yield needed insights into the spread of cancer as a result of the level of detail that can be captured. In recent years, in silico methods have been developed that can accurately and efficiently model the circulatory flows of cancer and other biological cells. These computational methods are capable of resolving detailed fluid flow fields which transport cells through tortuous physiological geometries, as well as the deformation and interactions between cells, cell-to-endothelium interactions, and tumor cell aggregates, all of which play important roles in metastatic spread. Such models can provide a powerful complement to experimental works, and a promising approach to recapitulating the endogenous setting while maintaining control over parameters such as shear rate, cell deformability, and the strength of adhesive binding to better understand tumor cell transport. In this review, we present an overview of computational models that have been developed for modeling cancer cells in the microcirculation, including insights they have provided into cell transport phenomena.
Collapse
Affiliation(s)
- Daniel F Puleri
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Peter Balogh
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA
| | - Amanda Randles
- Department of Biomedical Engineering, Duke University, Durham, NC, 27708, USA.
| |
Collapse
|
32
|
Li Y, Konstantopoulos K, Zhao R, Mori Y, Sun SX. The importance of water and hydraulic pressure in cell dynamics. J Cell Sci 2020; 133:133/20/jcs240341. [PMID: 33087485 DOI: 10.1242/jcs.240341] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
All mammalian cells live in the aqueous medium, yet for many cell biologists, water is a passive arena in which proteins are the leading players that carry out essential biological functions. Recent studies, as well as decades of previous work, have accumulated evidence to show that this is not the complete picture. Active fluxes of water and solutes of water can play essential roles during cell shape changes, cell motility and tissue function, and can generate significant mechanical forces. Moreover, the extracellular resistance to water flow, known as the hydraulic resistance, and external hydraulic pressures are important mechanical modulators of cell polarization and motility. For the cell to maintain a consistent chemical environment in the cytoplasm, there must exist an intricate molecular system that actively controls the cell water content as well as the cytoplasmic ionic content. This system is difficult to study and poorly understood, but ramifications of which may impact all aspects of cell biology from growth to metabolism to development. In this Review, we describe how mammalian cells maintain the cytoplasmic water content and how water flows across the cell surface to drive cell movement. The roles of mechanical forces and hydraulic pressure during water movement are explored.
Collapse
Affiliation(s)
- Yizeng Li
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.,Department of Mechanical Engineering, Kennesaw State University. Marietta, GA 30060, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.,Institute of NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Runchen Zhao
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.,Institute of NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yoichiro Mori
- Department of Mathematics and Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sean X Sun
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA .,Institute of NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA.,Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
33
|
Guerrero PE, Miró L, Wong BS, Massaguer A, Martínez-Bosch N, de Llorens R, Navarro P, Konstantopoulos K, Llop E, Peracaula R. Knockdown of α2,3-Sialyltransferases Impairs Pancreatic Cancer Cell Migration, Invasion and E-selectin-Dependent Adhesion. Int J Mol Sci 2020; 21:ijms21176239. [PMID: 32872308 PMCID: PMC7503936 DOI: 10.3390/ijms21176239] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/26/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Aberrant sialylation is frequently found in pancreatic ductal adenocarcinoma (PDA). α2,3-Sialyltransferases (α2,3-STs) ST3GAL3 and ST3GAL4 are overexpressed in PDA tissues and are responsible for increased biosynthesis of sialyl-Lewis (sLe) antigens, which play an important role in metastasis. This study addresses the effect of α2,3-STs knockdown on the migratory and invasive phenotype of PDA cells, and on E-selectin-dependent adhesion. Characterization of the cell sialome, the α2,3-STs and fucosyltransferases involved in the biosynthesis of sLe antigens, using a panel of human PDA cells showed differences in the levels of sialylated determinants and α2,3-STs expression, reflecting their phenotypic heterogeneity. Knockdown of ST3GAL3 and ST3GAL4 in BxPC-3 and Capan-1 cells, which expressed moderate to high levels of sLe antigens and α2,3-STs, led to a significant reduction in sLex and in most cases in sLea, with slight increases in the α2,6-sialic acid content. Moreover, ST3GAL3 and ST3GAL4 downregulation resulted in a significant decrease in cell migration and invasion. Binding and rolling to E-selectin, which represent key steps in metastasis, were also markedly impaired in the α2,3-STs knockdown cells. Our results indicate that inhibition of ST3GAL3 and ST3GAL4 may be a novel strategy to block PDA metastasis, which is one of the reasons for its dismal prognosis.
Collapse
Affiliation(s)
- Pedro Enrique Guerrero
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, 17003 Girona, Spain; (P.E.G.); (L.M.); (A.M.); (R.d.L.)
| | - Laura Miró
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, 17003 Girona, Spain; (P.E.G.); (L.M.); (A.M.); (R.d.L.)
| | - Bin S. Wong
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; (B.S.W.); (K.K.)
| | - Anna Massaguer
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, 17003 Girona, Spain; (P.E.G.); (L.M.); (A.M.); (R.d.L.)
| | - Neus Martínez-Bosch
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, 08003 Barcelona, Spain; (N.M.-B.); (P.N.)
| | - Rafael de Llorens
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, 17003 Girona, Spain; (P.E.G.); (L.M.); (A.M.); (R.d.L.)
| | - Pilar Navarro
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, 08003 Barcelona, Spain; (N.M.-B.); (P.N.)
- Institute of Biomedical Research of Barcelona (IIBB)-CSIC, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; (B.S.W.); (K.K.)
| | - Esther Llop
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, 17003 Girona, Spain; (P.E.G.); (L.M.); (A.M.); (R.d.L.)
- Correspondence: (E.L.); (R.P.); Tel.: +972-418370 (R.P.); Fax: +972-41-82-41 (R.P.)
| | - Rosa Peracaula
- Department of Biology, Biochemistry and Molecular Biology Unit, University of Girona, 17003 Girona, Spain; (P.E.G.); (L.M.); (A.M.); (R.d.L.)
- Correspondence: (E.L.); (R.P.); Tel.: +972-418370 (R.P.); Fax: +972-41-82-41 (R.P.)
| |
Collapse
|
34
|
SPARCL1 Influences Bovine Skeletal Muscle-Derived Satellite Cell Migration and Differentiation through an ITGB1-Mediated Signaling Pathway. Animals (Basel) 2020; 10:ani10081361. [PMID: 32781616 PMCID: PMC7460340 DOI: 10.3390/ani10081361] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/01/2020] [Accepted: 08/04/2020] [Indexed: 12/27/2022] Open
Abstract
Simple Summary It is known that cell migration and differentiation have a very important yet simple basis for muscle development and muscle disease treatment. Secreted protein acidic and rich in cysteine like 1 (SPARCL1), one of the components of extracellular matrix, has been proved to regulate bovine skeletal muscle-derived satellite cell differentiation. However, the exact mechanism is not yet clear. This study reveals that SPARCL1 promotes muscle-derived satellite cell early differentiation through integrin β1, thereby providing a new insight into the role of SPARCL1 in muscle development. Abstract As an extracellular matrix protein, secreted protein acidic and rich in cysteine (SPARC)-like 1 (SPARCL1) is involved in various cell functions. It was previously implicated in bovine skeletal muscle-derived satellite cell (MDSC) differentiation; however, the underlying mechanism remains unknown. In this study, immunoprecipitation and mass spectrometry revealed that integrin β1 (ITGB1) combines with SPARCL1. Further, co-immunoprecipitation demonstrated that SPARCL1 interacts with ITGB1. Cell scratch assays explored the influence of SPARCL1 on MDSC migration through ITGB1. In addition, desmin staining for myotube fusion rate and MyoD protein expression results showed that SPARCL1 promotes MDSC early differentiation through ITGB1. Furthermore, Western blotting results demonstrated that SPARCL1 regulates the expression of p-FAK, p-paxillin, vinculin, Cdc42, and Arp2/3 through ITGB1. These findings indicate that SPARCL1 may influence bovine MDSC migration and differentiation through an ITGB1-mediated cell signaling pathway. Herein, we elucidated the mechanism through which SPARCL1 affects MDSC differentiation. Our results provide insight into the molecular mechanism of muscle development and may in the future facilitate skeletal muscle regeneration and treatment.
Collapse
|
35
|
Wisniewski EO, Mistriotis P, Bera K, Law RA, Zhang J, Nikolic M, Weiger M, Parlani M, Tuntithavornwat S, Afthinos A, Zhao R, Wirtz D, Kalab P, Scarcelli G, Friedl P, Konstantopoulos K. Dorsoventral polarity directs cell responses to migration track geometries. SCIENCE ADVANCES 2020; 6:eaba6505. [PMID: 32789173 PMCID: PMC7399493 DOI: 10.1126/sciadv.aba6505] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 06/12/2020] [Indexed: 05/02/2023]
Abstract
How migrating cells differentially adapt and respond to extracellular track geometries remains unknown. Using intravital imaging, we demonstrate that invading cells exhibit dorsoventral (top-to-bottom) polarity in vivo. To investigate the impact of dorsoventral polarity on cell locomotion through different confining geometries, we fabricated microchannels of fixed cross-sectional area, albeit with distinct aspect ratios. Vertical confinement, exerted along the dorsoventral polarity axis, induces myosin II-dependent nuclear stiffening, which results in RhoA hyperactivation at the cell poles and slow bleb-based migration. In lateral confinement, directed perpendicularly to the dorsoventral polarity axis, the absence of perinuclear myosin II fails to increase nuclear stiffness. Hence, cells maintain basal RhoA activity and display faster mesenchymal migration. In summary, by integrating microfabrication, imaging techniques, and intravital microscopy, we demonstrate that dorsoventral polarity, observed in vivo and in vitro, directs cell responses in confinement by spatially tuning RhoA activity, which controls bleb-based versus mesenchymal migration.
Collapse
Affiliation(s)
- Emily O. Wisniewski
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Panagiotis Mistriotis
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Kaustav Bera
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Robert A. Law
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jitao Zhang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Milos Nikolic
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Maryland Biophysics Program, Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742, USA
| | - Michael Weiger
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maria Parlani
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Soontorn Tuntithavornwat
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Alexandros Afthinos
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Runchen Zhao
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Physical Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Petr Kalab
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Giuliano Scarcelli
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Peter Friedl
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Cancer Genomics Centre, 3584 Utrecht, Netherlands
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Physical Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
36
|
Liu Z, Lee SJ, Park S, Konstantopoulos K, Glunde K, Chen Y, Barman I. Cancer cells display increased migration and deformability in pace with metastatic progression. FASEB J 2020; 34:9307-9315. [PMID: 32463148 PMCID: PMC7547847 DOI: 10.1096/fj.202000101rr] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/24/2022]
Abstract
In this study, we explored the relation between metastatic states vs the capacity of confined migration, amoeboid transition, and cellular stiffness. We compared across an isogenic panel of human breast cancer cells derived from MDA-MB-231 cells. It was observed that cells after lung metastasis have the fastest migration and lowest stiffness, with a significantly higher capacity to transition into an amoeboid mode. Our findings illustrate that metastasis is a selective process favoring motile and softer cells. Moreover, the observation that circulating tumor cells resemble the parental cell line, but not lung-metastatic cells, suggests that cells with higher deformability and motility are likely selected during extravasation and colonization.
Collapse
Affiliation(s)
- Zhenhui Liu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Se Jong Lee
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Seungman Park
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Kristine Glunde
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yun Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD, USA
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD, USA
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
37
|
Vaeyens MM, Jorge-Peñas A, Barrasa-Fano J, Shapeti A, Roeffaers M, Van Oosterwyck H. Actomyosin-dependent invasion of endothelial sprouts in collagen. Cytoskeleton (Hoboken) 2020; 77:261-276. [PMID: 32588525 DOI: 10.1002/cm.21624] [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] [Received: 04/16/2020] [Revised: 06/11/2020] [Accepted: 06/22/2020] [Indexed: 12/30/2022]
Abstract
During sprouting angiogenesis-the growth of blood vessels from the existing vasculature-endothelial cells (ECs) adopt an elongated invasive form and exert forces at cell-cell and cell-matrix interaction sites. These cell shape changes and cellular tractions require extensive reorganizations of the actomyosin network. However, the respective roles of actin and myosin for endothelial sprouting are not fully elucidated. In this study, we further investigate these roles by treating 2D-migrating and 3D-sprouting ECs with chemical compounds targeting either myosin or actin. These treatments affected the endothelial cytoskeleton drastically and reduced the invasive response in a compound-specific manner; pointing toward a tight control of the actin and myosin activity during sprouting. Clusters in the data further illustrate that endothelial sprout morphology is sensitive to the in vitro model mechanical microenvironment and directs future research toward mechanical substrate guidance as a strategy for promoting engineered tissue vascularization. In summary, our results add to a growing corpus of research highlighting a key role of the cytoskeleton for sprouting angiogenesis.
Collapse
Affiliation(s)
- Marie-Mo Vaeyens
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Alvaro Jorge-Peñas
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Jorge Barrasa-Fano
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Apeksha Shapeti
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Maarten Roeffaers
- Department of Microbial and Molecular Systems (M2S), Centre for Membrane Separations, Adsorption, Catalysis and Spectroscopy for Sustainable Solutions (cMACS), KU Leuven, Leuven, Belgium
| | - Hans Van Oosterwyck
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
38
|
Griffith CM, Huang SA, Cho C, Khare TM, Rich M, Lee GH, Ligler FS, Diekman BO, Polacheck WJ. Microfluidics for the study of mechanotransduction. JOURNAL OF PHYSICS D: APPLIED PHYSICS 2020; 53:224004. [PMID: 33840837 PMCID: PMC8034607 DOI: 10.1088/1361-6463/ab78d4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Mechanical forces regulate a diverse set of biological processes at cellular, tissue, and organismal length scales. Investigating the cellular and molecular mechanisms that underlie the conversion of mechanical forces to biological responses is challenged by limitations of traditional animal models and in vitro cell culture, including poor control over applied force and highly artificial cell culture environments. Recent advances in fabrication methods and material processing have enabled the development of microfluidic platforms that provide precise control over the mechanical microenvironment of cultured cells. These devices and systems have proven to be powerful for uncovering and defining mechanisms of mechanotransduction. In this review, we first give an overview of the main mechanotransduction pathways that function at sites of cell adhesion, many of which have been investigated with microfluidics. We then discuss how distinct microfluidic fabrication methods can be harnessed to gain biological insight, with description of both monolithic and replica molding approaches. Finally, we present examples of how microfluidics can be used to apply both solid forces (substrate mechanics, strain, and compression) and fluid forces (luminal, interstitial) to cells. Throughout the review, we emphasize the advantages and disadvantages of different fabrication methods and applications of force in order to provide perspective to investigators looking to apply forces to cells in their own research.
Collapse
Affiliation(s)
- Christian M Griffith
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Stephanie A Huang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Crescentia Cho
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Tanmay M Khare
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC
| | - Matthew Rich
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - Gi-Hun Lee
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Frances S Ligler
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Brian O Diekman
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
- McAllister Heart Institute, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
- Cancer Cell Biology Program, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| |
Collapse
|
39
|
Doolin MT, Moriarty RA, Stroka KM. Mechanosensing of Mechanical Confinement by Mesenchymal-Like Cells. Front Physiol 2020; 11:365. [PMID: 32390868 PMCID: PMC7193100 DOI: 10.3389/fphys.2020.00365] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) and tumor cells have the unique capability to migrate out of their native environment and either home or metastasize, respectively, through extremely heterogeneous environments to a distant location. Once there, they can either aid in tissue regrowth or impart an immunomodulatory effect in the case of MSCs, or form secondary tumors in the case of tumor cells. During these journeys, cells experience physically confining forces that impinge on the cell body and the nucleus, ultimately causing a multitude of cellular changes. Most drastically, confining individual MSCs within hydrogels or confining monolayers of MSCs within agarose wells can sway MSC lineage commitment, while applying a confining compressive stress to metastatic tumor cells can increase their invasiveness. In this review, we seek to understand the signaling cascades that occur as cells sense confining forces and how that translates to behavioral changes, including elongated and multinucleated cell morphologies, novel migrational mechanisms, and altered gene expression, leading to a unique MSC secretome that could hold great promise for anti-inflammatory treatments. Through comparison of these altered behaviors, we aim to discern how MSCs alter their lineage selection, while tumor cells may become more aggressive and invasive. Synthesizing this information can be useful for employing MSCs for therapeutic approaches through systemic injections or tissue engineered grafts, and developing improved strategies for metastatic cancer therapies.
Collapse
Affiliation(s)
- Mary T. Doolin
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD, United States
| | - Rebecca A. Moriarty
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD, United States
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD, United States
- Maryland Biophysics Program, University of Maryland, College Park, College Park, MD, United States
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD, United States
| |
Collapse
|
40
|
Loisy A, Eggers J, Liverpool TB. How many ways a cell can move: the modes of self-propulsion of an active drop. SOFT MATTER 2020; 16:3106-3124. [PMID: 32154549 DOI: 10.1039/d0sm00070a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Numerous physical models have been proposed to explain how cell motility emerges from internal activity, mostly focused on how crawling motion arises from internal processes. Here we offer a classification of self-propulsion mechanisms based on general physical principles, showing that crawling is not the only way for cells to move on a substrate. We consider a thin drop of active matter on a planar substrate and fully characterize its autonomous motion for all three possible sources of driving: (i) the stresses induced in the bulk by active components, which allow in particular tractionless motion, (ii) the self-propulsion of active components at the substrate, which gives rise to crawling motion, and (iii) a net capillary force, possibly self-generated, and coupled to internal activity. We determine travelling-wave solutions to the lubrication equations as a function of a dimensionless activity parameter for each mode of motion. Numerical simulations are used to characterize the drop motion over a wide range of activity magnitudes, and explicit analytical solutions in excellent agreement with the simulations are derived in the weak-activity regime.
Collapse
Affiliation(s)
- Aurore Loisy
- School of Mathematics, University of Bristol, Bristol BS8 1UG, UK.
| | | | | |
Collapse
|
41
|
Zhong J, Yang Y, Liao L, Zhang C. Matrix stiffness-regulated cellular functions under different dimensionalities. Biomater Sci 2020; 8:2734-2755. [DOI: 10.1039/c9bm01809c] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The microenvironments that cells encounter with in vitro.
Collapse
Affiliation(s)
- Jiajun Zhong
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instruments (Sun Yat-sen University)
- School of Biomedical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Yuexiong Yang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instruments (Sun Yat-sen University)
- School of Biomedical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Liqiong Liao
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering
- Biomaterials Research Center
- School of Biomedical Engineering
- Southern Medical University
- Guangzhou
| | - Chao Zhang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instruments (Sun Yat-sen University)
- School of Biomedical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| |
Collapse
|
42
|
Padhi A, Thomson AH, Perry JB, Davis GN, McMillan RP, Loesgen S, Kaweesa EN, Kapania R, Nain AS, Brown DA. Bioenergetics underlying single-cell migration on aligned nanofiber scaffolds. Am J Physiol Cell Physiol 2019; 318:C476-C485. [PMID: 31875698 DOI: 10.1152/ajpcell.00221.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cell migration is centrally involved in a myriad of physiological processes, including morphogenesis, wound healing, tissue repair, and metastatic growth. The bioenergetics that underlie migratory behavior are not fully understood, in part because of variations in cell culture media and utilization of experimental cell culture systems that do not model physiological connective extracellular fibrous networks. In this study, we evaluated the bioenergetics of C2C12 myoblast migration and force production on fibronectin-coated nanofiber scaffolds of controlled diameter and alignment, fabricated using a nonelectrospinning spinneret-based tunable engineered parameters (STEP) platform. The contribution of various metabolic pathways to cellular migration was determined using inhibitors of cellular respiration, ATP synthesis, glycolysis, or glucose uptake. Despite immediate effects on oxygen consumption, mitochondrial inhibition only modestly reduced cell migration velocity, whereas inhibitors of glycolysis and cellular glucose uptake led to striking decreases in migration. The migratory metabolic sensitivity was modifiable based on the substrates present in cell culture media. Cells cultured in galactose (instead of glucose) showed substantial migratory sensitivity to mitochondrial inhibition. We used nanonet force microscopy to determine the bioenergetic factors responsible for single-cell force production and observed that neither mitochondrial nor glycolytic inhibition altered single-cell force production. These data suggest that myoblast migration is heavily reliant on glycolysis in cells grown in conventional media. These studies have wide-ranging implications for the causes, consequences, and putative therapeutic treatments aimed at cellular migration.
Collapse
Affiliation(s)
- Abinash Padhi
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia
| | - Alexander H Thomson
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia
| | - Justin B Perry
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia
| | - Grace N Davis
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia
| | - Ryan P McMillan
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia.,Virginia Tech Metabolism Core, Blacksburg, Virginia
| | - Sandra Loesgen
- Department of Chemistry, Oregon State University, Corvallis, Oregon
| | | | - Rakesh Kapania
- Department of Aerospace and Ocean Engineering, Virginia Tech, Blacksburg, Virginia
| | - Amrinder S Nain
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia
| | - David A Brown
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia.,Virginia Tech Center for Drug Discovery, Blacksburg, Virginia.,Virginia Tech Metabolism Core, Blacksburg, Virginia
| |
Collapse
|
43
|
Loisy A, Eggers J, Liverpool TB. Tractionless Self-Propulsion of Active Drops. PHYSICAL REVIEW LETTERS 2019; 123:248006. [PMID: 31922859 DOI: 10.1103/physrevlett.123.248006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Indexed: 06/10/2023]
Abstract
We report on a new mode of self-propulsion exhibited by compact drops of active liquids on a substrate which, remarkably, is tractionless, i.e., which imparts no mechanical stress locally on the surface. We show, both analytically and by numerical simulation, that the equations of motion for an active nematic drop possess a simple self-propelling solution, with no traction on the solid surface and in which the direction of motion is controlled by the winding of the nematic director field across the drop height. The physics underlying this mode of motion has the same origins as that giving rise to the zero viscosity observed in bacterial suspensions. This topologically protected tractionless self-propusion provides a robust physical mechanism for efficient cell migration in crowded environments like tissues.
Collapse
Affiliation(s)
- Aurore Loisy
- School of Mathematics, University of Bristol, Bristol BS8 1UG, United Kingdom
| | - Jens Eggers
- School of Mathematics, University of Bristol, Bristol BS8 1UG, United Kingdom
| | | |
Collapse
|
44
|
Morrow CM, Mukherjee A, Traore MA, Leaman EJ, Kim A, Smith EM, Nain AS, Behkam B. Integrating nanofibers with biochemical gradients to investigate physiologically-relevant fibroblast chemotaxis. LAB ON A CHIP 2019; 19:3641-3651. [PMID: 31560021 DOI: 10.1039/c9lc00602h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Persistent cell migration can occur due to anisotropy in the extracellular matrix (ECM), the gradient of a chemo-effector, or a combination of both. Through a variety of in vitro platforms, the contributions of either stimulus have been extensively studied, while the combined effect of both cues remains poorly described. Here, we report an integrative microfluidic chemotaxis assay device that enables the study of single cell chemotaxis on ECM-mimicking, aligned, and suspended nanofibers. Using this assay, we evaluated the effect of fiber spacing on the morphology and chemotaxis response of embryonic murine NIH/3T3 fibroblasts in the presence of temporally invariant, linear gradients of platelet-derived growth factor-BB (PDGF-BB). We found that the strength of PDGF-mediated chemotaxis response depends on not only the gradient slope but also the cell morphology. Low aspect ratio (3.4 ± 0.2) cells on flat substrata exhibited a chemotaxis response only at a PDGF-BB gradient of 0-10 ng mL-1. However, high aspect ratio (19.1 ± 0.7) spindle-shaped cells attached to individual fibers exhibited maximal chemotaxis response at a ten-fold shallower gradient of 0-1 ng mL-1, which was robustly maintained up to 0-10 ng mL-1. Quadrilateral-shaped cells of intermediate aspect ratio (13.6 ± 0.8) attached to two fibers exhibited a weaker response compared to the spindle-shaped cells, but still stronger compared to cells attached to 2D featureless substrata. Through pharmacological inhibition, we show that the mesenchymal chemotaxis pathway is conserved in cells on fibers. Altogether, our findings show that chemotaxis on ECM-mimicking fibers is modulated by fiber spacing-driven cell shape and can be significantly different from the behavior observed on flat 2D substrata. We envisage that this microfluidic platform will have wide applicability in understanding the combined role of ECM architecture and chemotaxis in physiological and pathological processes.
Collapse
Affiliation(s)
- Carmen M Morrow
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Apratim Mukherjee
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Mahama A Traore
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA. and School of Biomedical Engineering & Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Eric J Leaman
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA.
| | - AhRam Kim
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Evan M Smith
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA.
| | - Amrinder S Nain
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA. and School of Biomedical Engineering & Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Bahareh Behkam
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA. and School of Biomedical Engineering & Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
45
|
Doolin MT, Stroka KM. Integration of Mesenchymal Stem Cells into a Novel Micropillar Confinement Assay. Tissue Eng Part C Methods 2019; 25:662-676. [PMID: 31347455 PMCID: PMC6998058 DOI: 10.1089/ten.tec.2019.0083] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/24/2019] [Indexed: 01/12/2023] Open
Abstract
Mechanical cues such as stiffness have been shown to influence cell gene expression, protein expression, and cell behaviors critical for tissue engineering. The mechanical cue of confinement is also a pervasive parameter affecting cells in vivo and in tissue-engineered constructs. Despite its prevalence, the mechanical cue of confinement lacks assays that provide precise control over the degree of confinement induced on cells, yield a large sample size, enable long-term culture, and enable easy visualization of cells over time. In this study, we developed a process to systematically confine cells using micropillar arrays. Using photolithography and polydimethylsiloxane (PDMS) molding, we created PDMS arrays of micropillars that were 5, 10, 20, or 50 μm in spacing and between 13 and 17 μm in height. The tops of micropillars were coated with Pluronic F127 to inhibit cell adhesion, and we observed that mesenchymal stem cells (MSCs) robustly infiltrated into the micropillar arrays. MSC and nucleus morphology were altered by narrowing the micropillar spacing, and cytoskeletal elements within MSCs appeared to become more diffuse with increasing confinement. Specifically, MSCs exhibited a ring of actin around their periphery and punctate focal adhesions. MSC migration speed was reduced by narrowing micropillar spacing, and distinct migration behaviors of MSCs emerged in the presence of micropillars. MSCs continued to proliferate within micropillar arrays after 3 weeks in culture, displaying our assay's capability for long-term studies. Our assay also has the capacity to provide adequate cell numbers for quantitative assays to investigate the effect of confinement on gene and protein expression. Through deeper understanding of cell mechanotransduction in the context of confinement, we can modify tissue-engineered constructs to be optimal for a given purpose. Impact Statement In this study, we developed a novel process to systematically confine cells using micropillar arrays. Our assay provides insight into cell behavior in response to mechanical confinement. Through deeper understanding of how cells sense and respond to confinement, we can fine tune tissue-engineered constructs to be optimal for a given purpose. By combining confinement with other physical cues, we can harness mechanical properties to encourage or inhibit cell migration, direct cells down a particular lineage, induce cell secretion of specific cytokines or extracellular vesicles, and ultimately direct cells to behave in a way conducive to tissue engineering.
Collapse
Affiliation(s)
- Mary T. Doolin
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
- Biophysics Program, University of Maryland, College Park, Maryland
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, Maryland
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| |
Collapse
|
46
|
Jana A, Nookaew I, Singh J, Behkam B, Franco AT, Nain AS. Crosshatch nanofiber networks of tunable interfiber spacing induce plasticity in cell migration and cytoskeletal response. FASEB J 2019; 33:10618-10632. [PMID: 31225977 PMCID: PMC6766658 DOI: 10.1096/fj.201900131r] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/30/2019] [Indexed: 01/14/2023]
Abstract
Biomechanical cues within tissue microenvironments are critical for maintaining homeostasis, and their disruption can contribute to malignant transformation and metastasis. Once transformed, metastatic cancer cells can migrate persistently by adapting (plasticity) to changes in the local fibrous extracellular matrix, and current strategies to recapitulate persistent migration rely exclusively on the use of aligned geometries. Here, the controlled interfiber spacing in suspended crosshatch networks of nanofibers induces cells to exhibit plasticity in migratory behavior (persistent and random) and the associated cytoskeletal arrangement. At dense spacing (3 and 6 µm), unexpectedly, elongated cells migrate persistently (in 1 dimension) at high speeds in 3-dimensional shapes with thick nuclei, and short focal adhesion cluster (FAC) lengths. With increased spacing (18 and 36 µm), cells attain 2-dimensional morphologies, have flattened nuclei and longer FACs, and migrate randomly by rapidly detaching their trailing edges that strain the nuclei by ∼35%. At 54-µm spacing, kite-shaped cells become near stationary. Poorly developed filamentous actin stress fibers are found only in cells on 3-µm networks. Gene-expression profiling shows a decrease in transcriptional potential and a differential up-regulation of metabolic pathways. The consistency in observed phenotypes across cell lines supports using this platform to dissect hallmarks of plasticity in migration in vitro.-Jana, A., Nookaew, I., Singh, J., Behkam, B., Franco, A. T., Nain, A. S. Crosshatch nanofiber networks of tunable interfiber spacing induce plasticity in cell migration and cytoskeletal response.
Collapse
Affiliation(s)
- Aniket Jana
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Intawat Nookaew
- Department of Physiology and Biophysics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jugroop Singh
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Bahareh Behkam
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Aime T. Franco
- Department of Physiology and Biophysics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Amrinder S. Nain
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| |
Collapse
|
47
|
Mosier JA, Rahman-Zaman A, Zanotelli MR, VanderBurgh JA, Bordeleau F, Hoffman BD, Reinhart-King CA. Extent of Cell Confinement in Microtracks Affects Speed and Results in Differential Matrix Strains. Biophys J 2019; 117:1692-1701. [PMID: 31623884 DOI: 10.1016/j.bpj.2019.09.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 08/15/2019] [Accepted: 09/18/2019] [Indexed: 12/27/2022] Open
Abstract
During metastasis, cancer cells navigate through a spatially heterogeneous extracellular matrix (ECM). Physical properties of ECM, including the degree of confinement, influence cell migration behavior. Here, utilizing in vitro three-dimensional collagen microtracks, we demonstrate that cell-ECM interactions, specifically the degree of spatial confinement, regulate migratory behavior. We found that cells migrate faster when they are fully confined, contacting all four walls (top, bottom, and two sides) of a collagen microtrack, compared with cells that are partially confined, contacting less than four walls. When fully confined, cells exhibit fewer but larger vinculin-containing adhesions and create greater strains in the surrounding matrix directed toward the cell body. In contrast, partially confined cells develop a more elongated morphology with smaller but significantly more vinculin-containing adhesions and displace the surrounding matrix less than fully confined cells. The resulting effect of increasing cell contractility via Rho activation is dependent on the number of walls with which the cell is in contact. Although matrix strains increase in both fully and partially confined cells, cells that are partially confined increase speed, whereas those in full confinement decrease speed. Together, these results suggest that the degree of cell-ECM contact during confined migration is a key determinant of speed, morphology, and cell-generated substrate strains during motility, and these factors may work in tandem to facilitate metastatic cell migration.
Collapse
Affiliation(s)
- Jenna A Mosier
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Aniqua Rahman-Zaman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Matthew R Zanotelli
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Jacob A VanderBurgh
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Francois Bordeleau
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Dép. Biologie Moléculaire, Biochimie Médicale et Pathologie, Université Laval, Québec, Canada
| | - Brenton D Hoffman
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Cynthia A Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York.
| |
Collapse
|
48
|
Energetic costs regulated by cell mechanics and confinement are predictive of migration path during decision-making. Nat Commun 2019; 10:4185. [PMID: 31519914 PMCID: PMC6744572 DOI: 10.1038/s41467-019-12155-z] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/15/2019] [Indexed: 02/07/2023] Open
Abstract
Cell migration during the invasion-metastasis cascade requires cancer cells to navigate a spatially complex microenvironment that presents directional choices to migrating cells. Here, we investigate cellular energetics during migration decision-making in confined spaces. Theoretical and experimental data show that energetic costs for migration through confined spaces are mediated by a balance between cell and matrix compliance as well as the degree of spatial confinement to direct decision-making. Energetic costs, driven by the cellular work needed to generate force for matrix displacement, increase with increasing cell stiffness, matrix stiffness, and degree of spatial confinement, limiting migration. By assessing energetic costs between possible migration paths, we can predict the probability of migration choice. Our findings indicate that motility in confined spaces imposes high energetic demands on migrating cells, and cells migrate in the direction of least confinement to minimize energetic costs. Therefore, therapeutically targeting metabolism may limit cancer cell migration and metastasis.
Collapse
|
49
|
Wang M, Cheng B, Yang Y, Liu H, Huang G, Han L, Li F, Xu F. Microchannel Stiffness and Confinement Jointly Induce the Mesenchymal-Amoeboid Transition of Cancer Cell Migration. NANO LETTERS 2019; 19:5949-5958. [PMID: 31414817 DOI: 10.1021/acs.nanolett.9b01597] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The physical confinement of cell microenvironment could enhance the invasive capability and drug resistance of cancer cells. However, due to the lack of in vitro experimental platform to mimic both stiffness and confinement of the tumor microenvironment, the underlying mechanism remains elusive. Here, we developed a hydrogel-based microchannel platform with independently tunable channel stiffness and width in a physiological range. We found that the migration speed of the cancer cell is influenced by the synergistic effect of channel stiffness and width. In addition, the mesenchymal-amoeboid transition has a strong correlation with the channel stiffness. Besides, with a developed computational model, the role of nuclear stiffness on cancer migration speed and thus the mesenchymal-amoeboid transition in microchannels was also revealed. This platform is capable of mimicking the native physical microenvironment during metastasis, providing a powerful tool for high-throughput screening applications and investigating the interaction between cancer migration and biophysical microenvironment.
Collapse
Affiliation(s)
| | | | | | | | | | - Lichun Han
- Department of Anesthesia , Xi'an Daxing Hospital , Xi'an 710049 , P.R. China
| | | | | |
Collapse
|
50
|
Park H, Doh J. T cell migration in microchannels densely packed with T cells. Sci Rep 2019; 9:7198. [PMID: 31076592 PMCID: PMC6510777 DOI: 10.1038/s41598-019-43569-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 04/23/2019] [Indexed: 01/22/2023] Open
Abstract
T cells migrate diverse microenvironments of the body to mount antigen-specific immune responses. T cell activation, a key initial process for antigen-specific immune responses, occur in secondary lymphoid organs such as spleens and lymph nodes where high density of T cells migrates rapidly through the reticular networks formed by stromal cells. In vitro model system recapitulating key characteristics of secondary lymphoid organs, confined spaces densely packed with rapidly migrating cells, would be useful to investigate mechanisms of T cell migration. In this study, we devised a method to fabricate microchannels densely packed with T cells. Microchannel arrays with fixed height (4 μm) and length (1.5 mm) and various widths (15~80 μm) were fabricated in between trapezoid-shaped reservoirs that facilitated T cell sedimentation near microchannel entries. Microchannel surface chemistry and filling time were optimized to achieve high packing density (0.89) of T cell filling within microchannels. Particle image velocimetry (PIV) analysis method was employed to extract velocity field of microchannels densely packed with T cells. Using velocity field information, various motility parameters were further evaluated to quantitatively assess the effects of microchannel width and media tonicity on T cell motility within cell dense microenvironments.
Collapse
Affiliation(s)
- HyoungJun Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH) San 31, Hyoja-dong, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea
| | - Junsang Doh
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH) San 31, Hyoja-dong, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea. .,School of Interdisciplinary Bioscience and Bioengineering (I-Bio), Pohang University of Science and Technology (POSTECH) San 31, Hyoja-dong, Nam-Gu, Pohang, Gyeongbuk, 37673, Korea. .,Department of Materials Science and Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.
| |
Collapse
|