1
|
Tao Y, Ghagre A, Molter CW, Clouvel A, Al Rahbani J, Brown CM, Nowrouzezahrai D, Ehrlicher AJ. Inferring cellular contractile forces and work using deep morphology traction microscopy. Biophys J 2024; 123:3217-3230. [PMID: 39033326 PMCID: PMC11427771 DOI: 10.1016/j.bpj.2024.07.020] [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: 07/19/2023] [Revised: 05/02/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024] Open
Abstract
Traction-force microscopy (TFM) has emerged as a widely used standard methodology to measure cell-generated traction forces and determine their role in regulating cell behavior. While TFM platforms have enabled many discoveries, their implementation remains limited due to complex experimental procedures, specialized substrates, and the ill-posed inverse problem whereby low-magnitude high-frequency noise in the displacement field severely contaminates the resulting traction measurements. Here, we introduce deep morphology traction microscopy (DeepMorphoTM), a deep-learning alternative to conventional TFM approaches. DeepMorphoTM first infers cell-induced substrate displacement solely from a sequence of cell shapes and subsequently computes cellular traction forces, thus avoiding the requirement of a specialized fiduciarily marked deformable substrate or force-free reference image. Rather, this technique drastically simplifies the overall experimental methodology, imaging, and analysis needed to conduct cell-contractility measurements. We demonstrate that DeepMorphoTM quantitatively matches conventional TFM results while offering stability against the biological variability in cell contractility for a given cell shape. Without high-frequency noise in the inferred displacement, DeepMorphoTM also resolves the ill-posedness of traction computation, increasing the consistency and accuracy of traction analysis. We demonstrate the accurate extrapolation across several cell types and substrate materials, suggesting robustness of the methodology. Accordingly, we present DeepMorphoTM as a capable yet simpler alternative to conventional TFM for characterizing cellular contractility in two dimensions.
Collapse
Affiliation(s)
- Yuanyuan Tao
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada; Department of Electrical and Computer Engineering, McGill University, Montreal, Quebec, Canada
| | - Ajinkya Ghagre
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Clayton W Molter
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Anna Clouvel
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Jalal Al Rahbani
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Claire M Brown
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada; Department of Physiology, McGill University, Montreal, Quebec, Canada; Advanced BioImaging Facility (ABIF), McGill University, Montreal, Quebec, Canada
| | - Derek Nowrouzezahrai
- Department of Electrical and Computer Engineering, McGill University, Montreal, Quebec, Canada
| | - Allen J Ehrlicher
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada; Department of Mechanical Engineering, McGill University, Montreal, Quebec, Canada; Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montreal, Quebec, Canada; Centre for Structural Biology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
2
|
Son DO, Benitez R, Diao L, Hinz B. How to Keep Myofibroblasts under Control: Culture of Mouse Skin Fibroblasts on Soft Substrates. J Invest Dermatol 2024; 144:1923-1934. [PMID: 39078357 DOI: 10.1016/j.jid.2024.05.033] [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: 02/12/2024] [Revised: 05/03/2024] [Accepted: 05/21/2024] [Indexed: 07/31/2024]
Abstract
During the physiological healing of skin wounds, fibroblasts recruited from the uninjured adjacent dermis and deeper subcutaneous fascia layers are transiently activated into myofibroblasts to first secrete and then contract collagen-rich extracellular matrix into a mechanically resistant scar. Scar tissue restores skin integrity after damage but comes at the expense of poor esthetics and loss of tissue function. Stiff scar matrix also mechanically activates various precursor cells into myofibroblasts in a positive feedback loop. Persistent myofibroblast activation results in pathologic accumulation of fibrous collagen and hypertrophic scarring, called fibrosis. Consequently, the mechanisms of fibroblast-to-myofibroblast activation and persistence are studied to develop antifibrotic and prohealing treatments. Mechanistic understanding often starts in a plastic cell culture dish. This can be problematic because contact of fibroblasts with tissue culture plastic or glass surfaces invariably generates myofibroblast phenotypes in standard culture. We describe a straight-forward method to produce soft cell culture surfaces for fibroblast isolation and continued culture and highlight key advantages and limitations of the approach. Adding a layer of elastic silicone polymer tunable to the softness of normal skin and the stiffness of pathologic scars allows to control mechanical fibroblast activation while preserving the simplicity of conventional 2-dimensional cell culture.
Collapse
Affiliation(s)
- Dong Ok Son
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | - Raquel Benitez
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | - Li Diao
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | - Boris Hinz
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada; Faculty of Dentistry, University of Toronto, Toronto, Canada.
| |
Collapse
|
3
|
Wagner R, Carré MJ, Perrault CM, Evans PC, Lewis R. Assessing friction and damage of cell monolayers on soft substrates in vitro. J R Soc Interface 2024; 21:20230696. [PMID: 38842440 DOI: 10.1098/rsif.2023.0696] [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: 11/27/2023] [Accepted: 03/11/2024] [Indexed: 06/07/2024] Open
Abstract
In the area of surgical applications, understanding the interaction between medical device materials and tissue is important since this interaction may cause complications. The interaction often consists of a cell monolayer touching the medical device that can be mimicked in vitro. Prominent examples of this are contact lenses, where epithelial cells interact with the contact lens, or stents and catheters, which are in contact with endothelial cells. To investigate those interactions, in previous studies, expensive microtribometers were used to avoid pressures in the contact area far beyond physiologically relevant levels. Here, we aim to present a new methodology that is cost- and time-efficient, more accessible than those used previously and allows for the application of more realistic pressures, while permitting a quantification of the damage caused to the monolayer. For this, a soft polydimethylsiloxane is employed that better mimics the mechanical properties of blood vessels than materials used in other studies. Furthermore, a technique to account for misalignments within the experiment set-up is presented. This is carried out using the raw spatial and force data recorded by the tribometer and adjusting for misalignments. The methodology is demonstrated using an endothelial cell (human umbilical vein endothelial cells) monolayer.
Collapse
Affiliation(s)
- Rasmus Wagner
- Department of Mechanical Engineering, University of Sheffield , Sheffield S1 3JD, UK
| | - Matt J Carré
- Department of Mechanical Engineering, University of Sheffield , Sheffield S1 3JD, UK
| | - Cecile M Perrault
- Department of Mechanical Engineering, University of Sheffield , Sheffield S1 3JD, UK
- Eden Microfluidics , Paris 75012, France
| | - Paul C Evans
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield , Sheffield S10 2RX, UK
| | - Roger Lewis
- Department of Mechanical Engineering, University of Sheffield , Sheffield S1 3JD, UK
| |
Collapse
|
4
|
Liang L, Song X, Zhao H, Lim CT. Insights into the mechanobiology of cancer metastasis via microfluidic technologies. APL Bioeng 2024; 8:021506. [PMID: 38841688 PMCID: PMC11151435 DOI: 10.1063/5.0195389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024] Open
Abstract
During cancer metastasis, cancer cells will encounter various microenvironments with diverse physical characteristics. Changes in these physical characteristics such as tension, stiffness, viscosity, compression, and fluid shear can generate biomechanical cues that affect cancer cells, dynamically influencing numerous pathophysiological mechanisms. For example, a dense extracellular matrix drives cancer cells to reorganize their cytoskeleton structures, facilitating confined migration, while this dense and restricted space also acts as a physical barrier that potentially results in nuclear rupture. Identifying these pathophysiological processes and understanding their underlying mechanobiological mechanisms can aid in the development of more effective therapeutics targeted to cancer metastasis. In this review, we outline the advances of engineering microfluidic devices in vitro and their role in replicating tumor microenvironment to mimic in vivo settings. We highlight the potential cellular mechanisms that mediate their ability to adapt to different microenvironments. Meanwhile, we also discuss some important mechanical cues that still remain challenging to replicate in current microfluidic devices in future direction. While much remains to be explored about cancer mechanobiology, we believe the developments of microfluidic devices will reveal how these physical cues impact the behaviors of cancer cells. It will be crucial in the understanding of cancer metastasis, and potentially contributing to better drug development and cancer therapy.
Collapse
Affiliation(s)
- Lanfeng Liang
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Xiao Song
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | | | | |
Collapse
|
5
|
Ghagre A, Delarue A, Srivastava LK, Koushki N, Ehrlicher A. Nuclear curvature determines Yes-associated protein localization and differentiation of mesenchymal stem cells. Biophys J 2024; 123:1222-1239. [PMID: 38605521 PMCID: PMC11140468 DOI: 10.1016/j.bpj.2024.04.008] [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: 07/19/2023] [Revised: 01/17/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024] Open
Abstract
Controlling mesenchymal stem cell (MSC) differentiation remains a critical challenge in MSCs' therapeutic application. Numerous biophysical and mechanical stimuli influence stem cell fate; however, their relative efficacy and specificity in mechanically directed differentiation remain unclear. Yes-associated protein (YAP) is one key mechanosensitive protein that controls MSC differentiation. Previous studies have related nuclear mechanics with YAP activity, but we still lack an understanding of what nuclear deformation specifically regulates YAP and its relationship with mechanical stimuli. Here, we report that maximum nuclear curvature is the most precise biophysical determinant for YAP mechanotransduction-mediated MSC differentiation and is a relevant parameter for stem cell-based therapies. We employed traction force microscopy and confocal microscopy to characterize the causal relationships between contractility and nuclear deformation in regulating YAP activity in MSCs. We observed that an increase in contractility compresses nuclei anisotropically, whereby the degree of asymmetric compression increased the bending curvature of the nuclear membrane. We then examined membrane curvature and tension using thin micropatterned adhesive substrate lines and an FRET-based tension sensor, revealing the direct role of curvature in YAP activity driven by both active and passive nuclear import. Finally, we employed micropatterned lines to control nuclear curvature and precisely direct MSC differentiation. This work illustrates that nuclear curvature subsumes other biophysical aspects to control YAP-mediated differentiation in MSCs and may provide a deterministic solution to some of the challenges in mesenchymal stem cell therapies.
Collapse
Affiliation(s)
- Ajinkya Ghagre
- Department of Bioengineering, McGill University, Montreal, Canada
| | - Alice Delarue
- Department of Bioengineering, McGill University, Montreal, Canada
| | | | - Newsha Koushki
- Department of Bioengineering, McGill University, Montreal, Canada
| | - Allen Ehrlicher
- Department of Bioengineering, McGill University, Montreal, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, Canada; Department of Biomedical Engineering, McGill University, Montreal, Canada; Department of Mechanical Engineering, McGill University, Montreal, Canada; Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montreal, Canada; Centre for Structural Biology, McGill University, Montreal, Canada.
| |
Collapse
|
6
|
Schuftan D, Kooh YKG, Guo J, Sun Y, Aryan L, Stottlemire B, Berkland C, Genin GM, Huebsch N. Dynamic control of contractile resistance to iPSC-derived micro-heart muscle arrays. J Biomed Mater Res A 2024; 112:534-548. [PMID: 37952251 PMCID: PMC10922390 DOI: 10.1002/jbm.a.37642] [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: 05/31/2023] [Revised: 09/25/2023] [Accepted: 10/24/2023] [Indexed: 11/14/2023]
Abstract
Many types of cardiovascular disease are linked to the mechanical forces placed on the heart. However, our understanding of how mechanical forces exactly affect the cellular biology of the heart remains incomplete. In vitro models based on cardiomyocytes derived from human induced pluripotent stem cells (iPSC-CM) enable researchers to develop medium to high-throughput systems to study cardiac mechanobiology at the cellular level. Previous models have been developed to enable the study of mechanical forces, such as cardiac afterload. However, most of these models require exogenous extracellular matrix (ECM) to form cardiac tissues. Recently, a system was developed to simulate changes in afterload by grafting ECM-free micro-heart muscle arrays to elastomeric substrates of discrete stiffnesses. In the present study, we extended this system by combining the elastomer-grafted tissue arrays with a magnetorheological elastomeric substrate. This system allows iPSC-CM based micro-heart muscle arrays to experience dynamic changes in contractile resistance to mimic dynamically altered afterload. Acute changes in substrate stiffness led to acute changes in the calcium dynamics and contractile forces, illustrating the system's ability to dynamically elicit changes in tissue mechanics by dynamically changing contractile resistance.
Collapse
Affiliation(s)
- David Schuftan
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yasaman Kargar Gaz Kooh
- Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jingxuan Guo
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yuwen Sun
- Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Lavanya Aryan
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Bryce Stottlemire
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
| | - Cory Berkland
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - Guy M. Genin
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- NSF Center for Engineering Mechanobiology, St. Louis, Missouri, USA
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- NSF Center for Engineering Mechanobiology, St. Louis, Missouri, USA
| |
Collapse
|
7
|
Alberici Delsin LE, Plutoni C, Clouvel A, Keil S, Marpeaux L, Elouassouli L, Khavari A, Ehrlicher AJ, Emery G. MAP4K4 regulates forces at cell-cell and cell-matrix adhesions to promote collective cell migration. Life Sci Alliance 2023; 6:e202302196. [PMID: 37369604 PMCID: PMC10300198 DOI: 10.26508/lsa.202302196] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Collective cell migration is not only important for development and tissue homeostasis but can also promote cancer metastasis. To migrate collectively, cells need to coordinate cellular extensions and retractions, adhesion sites dynamics, and forces generation and transmission. Nevertheless, the regulatory mechanisms coordinating these processes remain elusive. Using A431 carcinoma cells, we identify the kinase MAP4K4 as a central regulator of collective migration. We show that MAP4K4 inactivation blocks the migration of clusters, whereas its overexpression decreases cluster cohesion. MAP4K4 regulates protrusion and retraction dynamics, remodels the actomyosin cytoskeleton, and controls the stability of both cell-cell and cell-substrate adhesion. MAP4K4 promotes focal adhesion disassembly through the phosphorylation of the actin and plasma membrane crosslinker moesin but disassembles adherens junctions through a moesin-independent mechanism. By analyzing traction and intercellular forces, we found that MAP4K4 loss of function leads to a tensional disequilibrium throughout the cell cluster, increasing the traction forces and the tension loading at the cell-cell adhesions. Together, our results indicate that MAP4K4 activity is a key regulator of biomechanical forces at adhesion sites, promoting collective migration.
Collapse
Affiliation(s)
- Lara Elis Alberici Delsin
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer https://ror.org/0161xgx34 (IRIC), Université de Montréal, Montréal, Canada
| | - Cédric Plutoni
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer https://ror.org/0161xgx34 (IRIC), Université de Montréal, Montréal, Canada
| | - Anna Clouvel
- Department of Bioengineering, McGill University, Montreal, Canada
| | - Sarah Keil
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer https://ror.org/0161xgx34 (IRIC), Université de Montréal, Montréal, Canada
| | - Léa Marpeaux
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer https://ror.org/0161xgx34 (IRIC), Université de Montréal, Montréal, Canada
| | - Lina Elouassouli
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer https://ror.org/0161xgx34 (IRIC), Université de Montréal, Montréal, Canada
| | - Adele Khavari
- Department of Bioengineering, McGill University, Montreal, Canada
| | | | - Gregory Emery
- Vesicular Trafficking and Cell Signalling Research Unit, Institute for Research in Immunology and Cancer https://ror.org/0161xgx34 (IRIC), Université de Montréal, Montréal, Canada
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, Canada
| |
Collapse
|
8
|
Ayad NM, Lakins JN, Ghagre A, Ehrlicher AJ, Weaver VM. Tissue tension permits β-catenin phosphorylation to drive mesoderm specification in human embryonic stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.14.549074. [PMID: 37503095 PMCID: PMC10370032 DOI: 10.1101/2023.07.14.549074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The role of morphogenetic forces in cell fate specification is an area of intense interest. Our prior studies suggested that the development of high cell-cell tension in human embryonic stem cells (hESC) colonies permits the Src-mediated phosphorylation of junctional β-catenin that accelerates its release to potentiate Wnt-dependent signaling critical for initiating mesoderm specification. Using an ectopically expressed nonphosphorylatable mutant of β-catenin (Y654F), we now provide direct evidence that impeding tension-dependent Src-mediated β-catenin phosphorylation impedes the expression of Brachyury (T) and the epithelial-to-mesenchymal transition (EMT) necessary for mesoderm specification. Addition of exogenous Wnt3a or inhibiting GSK3β activity rescued mesoderm expression, emphasizing the importance of force dependent Wnt signaling in regulating mechanomorphogenesis. Our work provides a framework for understanding tension-dependent β-catenin/Wnt signaling in the self-organization of tissues during developmental processes including gastrulation.
Collapse
Affiliation(s)
- Nadia M.E. Ayad
- Graduate Program in Bioengineering, University of California, San Francisco and University of California Berkeley, San Francisco, CA 94143, USA; Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Johnathon N. Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ajinkya Ghagre
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Allen J. Ehrlicher
- Department of Bioengineering, Department of Anatomy and Cell Biology, Department of Biomedical Engineering, Department of Mechanical Engineering, Centre for Structural Biology, Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF Comprehensive Cancer Center, Helen Diller Family Cancer Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences, Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
9
|
Koushki N, Ghagre A, Srivastava LK, Molter C, Ehrlicher AJ. Nuclear compression regulates YAP spatiotemporal fluctuations in living cells. Proc Natl Acad Sci U S A 2023; 120:e2301285120. [PMID: 37399392 PMCID: PMC10334804 DOI: 10.1073/pnas.2301285120] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/04/2023] [Indexed: 07/05/2023] Open
Abstract
Yes-associated protein (YAP) is a key mechanotransduction protein in diverse physiological and pathological processes; however, a ubiquitous YAP activity regulatory mechanism in living cells has remained elusive. Here, we show that YAP nuclear translocation is highly dynamic during cell movement and is driven by nuclear compression arising from cell contractile work. We resolve the mechanistic role of cytoskeletal contractility in nuclear compression by manipulation of nuclear mechanics. Disrupting the linker of nucleoskeleton and cytoskeleton complex reduces nuclear compression for a given contractility and correspondingly decreases YAP localization. Conversely, decreasing nuclear stiffness via silencing of lamin A/C increases nuclear compression and YAP nuclear localization. Finally, using osmotic pressure, we demonstrated that nuclear compression even without active myosin or filamentous actin regulates YAP localization. The relationship between nuclear compression and YAP localization captures a universal mechanism for YAP regulation with broad implications in health and biology.
Collapse
Affiliation(s)
- Newsha Koushki
- Department of Bioengineering, McGill University, Montreal, QCH3A 0E9, Canada
| | - Ajinkya Ghagre
- Department of Bioengineering, McGill University, Montreal, QCH3A 0E9, Canada
| | | | - Clayton Molter
- Department of Bioengineering, McGill University, Montreal, QCH3A 0E9, Canada
| | - Allen J. Ehrlicher
- Department of Bioengineering, McGill University, Montreal, QCH3A 0E9, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QCH3A 0C7, Canada
- Department of Biomedical Engineering, McGill University, Montreal, QCH3A 2B4, Canada
- Department of Mechanical Engineering, McGill University, Montreal, QCH3A 0C3, Canada
- Centre for Structural Biology, McGill University, Montreal, QCH3G 0B1, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QCH3A 1A3, Canada
| |
Collapse
|
10
|
Kim JH, Schaible N, Hall JK, Bartolák-Suki E, Deng Y, Herrmann J, Sonnenberg A, Behrsing HP, Lutchen KR, Krishnan R, Suki B. Multiscale stiffness of human emphysematous precision cut lung slices. SCIENCE ADVANCES 2023; 9:eadf2535. [PMID: 37205750 PMCID: PMC10198632 DOI: 10.1126/sciadv.adf2535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 04/14/2023] [Indexed: 05/21/2023]
Abstract
Emphysema is a debilitating disease that remodels the lung leading to reduced tissue stiffness. Thus, understanding emphysema progression requires assessing lung stiffness at both the tissue and alveolar scales. Here, we introduce an approach to determine multiscale tissue stiffness and apply it to precision-cut lung slices (PCLS). First, we established a framework for measuring stiffness of thin, disk-like samples. We then designed a device to verify this concept and validated its measuring capabilities using known samples. Next, we compared healthy and emphysematous human PCLS and found that the latter was 50% softer. Through computational network modeling, we discovered that this reduced macroscopic tissue stiffness was due to both microscopic septal wall remodeling and structural deterioration. Lastly, through protein expression profiling, we identified a wide spectrum of enzymes that can drive septal wall remodeling, which, together with mechanical forces, lead to rupture and structural deterioration of the emphysematous lung parenchyma.
Collapse
Affiliation(s)
- Jae Hun Kim
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Mechanobiologix, LLC, Newton, MA, USA
| | - Niccole Schaible
- Mechanobiologix, LLC, Newton, MA, USA
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Joseph K. Hall
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | | | - Yuqing Deng
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Jacob Herrmann
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- University of Iowa, Iowa City, IA, USA
| | - Adam Sonnenberg
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | | | - Kenneth R. Lutchen
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Ramaswamy Krishnan
- Mechanobiologix, LLC, Newton, MA, USA
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Mechanobiologix, LLC, Newton, MA, USA
| |
Collapse
|
11
|
Morales IA, Boghdady CM, Campbell BE, Moraes C. Integrating mechanical sensor readouts into organ-on-a-chip platforms. Front Bioeng Biotechnol 2022; 10:1060895. [PMID: 36588933 PMCID: PMC9800895 DOI: 10.3389/fbioe.2022.1060895] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Organs-on-a-chip have emerged as next-generation tissue engineered models to accurately capture realistic human tissue behaviour, thereby addressing many of the challenges associated with using animal models in research. Mechanical features of the culture environment have emerged as being critically important in designing organs-on-a-chip, as they play important roles in both stimulating realistic tissue formation and function, as well as capturing integrative elements of homeostasis, tissue function, and tissue degeneration in response to external insult and injury. Despite the demonstrated impact of incorporating mechanical cues in these models, strategies to measure these mechanical tissue features in microfluidically-compatible formats directly on-chip are relatively limited. In this review, we first describe general microfluidically-compatible Organs-on-a-chip sensing strategies, and categorize these advances based on the specific advantages of incorporating them on-chip. We then consider foundational and recent advances in mechanical analysis techniques spanning cellular to tissue length scales; and discuss their integration into Organs-on-a-chips for more effective drug screening, disease modeling, and characterization of biological dynamics.
Collapse
Affiliation(s)
| | | | | | - Christopher Moraes
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada,Department of Chemical Engineering, McGill University, Montreal, QC, Canada,Department of Biomedical Engineering, McGill University, Montreal, QC, Canada,*Correspondence: Christopher Moraes,
| |
Collapse
|
12
|
Snelders M, Koedijk IH, Schirmer J, Mulleners O, van Leeuwen J, de Wagenaar NP, Bartulos O, Voskamp P, Braam S, Guttenberg Z, Danser AJ, Majoor-Krakauer D, Meijering E, van der Pluijm I, Essers J. Contraction pressure analysis using optical imaging in normal and MYBPC3-mutated hiPSC-derived cardiomyocytes grown on matrices with tunable stiffness. BIOMATERIALS AND BIOSYSTEMS 2022; 8:100068. [PMID: 36824378 PMCID: PMC9934435 DOI: 10.1016/j.bbiosy.2022.100068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/09/2022] [Accepted: 10/15/2022] [Indexed: 12/04/2022] Open
Abstract
Current in vivo disease models and analysis methods for cardiac drug development have been insufficient in providing accurate and reliable predictions of drug efficacy and safety. Here, we propose a custom optical flow-based analysis method to quantitatively measure recordings of contracting cardiomyocytes on polydimethylsiloxane (PDMS), compatible with medium-throughput systems. Movement of the PDMS was examined by covalently bound fluorescent beads on the PDMS surface, differences caused by increased substrate stiffness were compared, and cells were stimulated with β-agonist. We further validated the system using cardiomyocytes treated with endothelin-1 and compared their contractions against control and cells incubated with receptor antagonist bosentan. After validation we examined two MYBPC3-mutant patient-derived cell lines. Recordings showed that higher substrate stiffness resulted in higher contractile pressure, while beating frequency remained similar to control. β-agonist stimulation resulted in both higher beating frequency as well as higher pressure values during contraction and relaxation. Cells treated with endothelin-1 showed an increased beating frequency, but a lower contraction pressure. Cells treated with both endothelin-1 and bosentan remained at control level of beating frequency and pressure. Lastly, both MYBPC3-mutant lines showed a higher beating frequency and lower contraction pressure. Our validated method is capable of automatically quantifying contraction of hiPSC-derived cardiomyocytes on a PDMS substrate of known shear modulus, returning an absolute value. Our method could have major benefits in a medium-throughput setting.
Collapse
Affiliation(s)
- Matthijs Snelders
- Department of Molecular Genetics, Erasmus MC, Rotterdam, the Netherlands
| | - Iris H. Koedijk
- Department of Molecular Genetics, Erasmus MC, Rotterdam, the Netherlands
| | | | - Otto Mulleners
- Department of Molecular Genetics, Erasmus MC, Rotterdam, the Netherlands
| | | | - Nathalie P. de Wagenaar
- Department of Molecular Genetics, Erasmus MC, Rotterdam, the Netherlands,Department of Cardiology, Erasmus MC, Rotterdam, the Netherlands
| | | | | | | | | | - A.H. Jan Danser
- Department of Internal Medicine - Pharmacology, Erasmus MC, Rotterdam, the Netherlands
| | | | - Erik Meijering
- School of Computer Science and Engineering, University of New South Wales, Sydney, Australia
| | - Ingrid van der Pluijm
- Department of Molecular Genetics, Erasmus MC, Rotterdam, the Netherlands,Department of Vascular Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Jeroen Essers
- Department of Molecular Genetics, Erasmus MC, Rotterdam, the Netherlands,Department of Vascular Surgery, Erasmus MC, Rotterdam, the Netherlands,Department of Radiotherapy, Erasmus MC, Rotterdam, the Netherlands,Corresponding author: Erasmus Medical Center, Wytemaweg 80, Rotterdam 3015CN, The Netherlands
| |
Collapse
|
13
|
Afthinos A, Bera K, Chen J, Ozcelikkale A, Amitrano A, Choudhury MI, Huang R, Pachidis P, Mistriotis P, Chen Y, Konstantopoulos K. Migration and 3D Traction Force Measurements inside Compliant Microchannels. NANO LETTERS 2022; 22:7318-7327. [PMID: 36112517 PMCID: PMC9872269 DOI: 10.1021/acs.nanolett.2c01261] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Cells migrate in vivo through channel-like tracks. While polydimethylsiloxane devices emulate such tracks in vitro, their channel walls are impermeable and have supraphysiological stiffness. Existing hydrogel-based platforms address these issues but cannot provide high-throughput analysis of cell motility in independently controllable stiffness and confinement. We herein develop polyacrylamide (PA)-based microchannels of physiological stiffness and prescribed dimensions for high-throughput analysis of cell migration and identify a biphasic dependence of speed upon confinement and stiffness. By utilizing novel four-walled microchannels with heterogeneous stiffness, we reveal the distinct contributions of apicolateral versus basal microchannel wall stiffness to confined versus unconfined migration. While the basal wall stiffness dictates unconfined migration, apicolateral stiffness controls confined migration. By tracking nanobeads embedded within channel walls, we innovate three-dimensional traction force measurements around spatially confining cells at subcellular resolution. Our unique and highly customizable device fabrication strategy provides a physiologically relevant in vitro platform to study confined cells.
Collapse
Affiliation(s)
- Alexandros Afthinos
- 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
| | - Junjie Chen
- 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
- Center for Cell Dynamics, The Johns Hopkins University, Baltimore MD, 21205, USA
| | - Altug Ozcelikkale
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore MD, 21218, USA
- Department of Mechanical Engineering, Middle East Technical University, 06531 Ankara, Turkey
| | - Alice Amitrano
- 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
| | - Mohammad Ikbal Choudhury
- 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
| | - Randy Huang
- 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
- Center for Cell Dynamics, The Johns Hopkins University, Baltimore MD, 21205, USA
| | - Pavlos Pachidis
- 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
| | - Yun Chen
- 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
- Center for Cell Dynamics, The Johns Hopkins University, Baltimore MD, 21205, 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 School of Medicine, Baltimore MD, 21205, USA
| |
Collapse
|
14
|
Molter CW, Muszynski EF, Tao Y, Trivedi T, Clouvel A, Ehrlicher AJ. Prostate cancer cells of increasing metastatic potential exhibit diverse contractile forces, cell stiffness, and motility in a microenvironment stiffness-dependent manner. Front Cell Dev Biol 2022; 10:932510. [PMID: 36200037 PMCID: PMC9527313 DOI: 10.3389/fcell.2022.932510] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
During metastasis, all cancer types must migrate through crowded multicellular environments. Simultaneously, cancers appear to change their biophysical properties. Indeed, cell softening and increased contractility are emerging as seemingly ubiquitous biomarkers of metastatic progression which may facilitate metastasis. Cell stiffness and contractility are also influenced by the microenvironment. Stiffer matrices resembling the tumor microenvironment cause metastatic cells to contract more strongly, further promoting contractile tumorigenic phenotypes. Prostate cancer (PCa), however, appears to deviate from these common cancer biophysics trends; aggressive metastatic PCa cells appear stiffer, rather than softer, to their lowly metastatic PCa counterparts. Although metastatic PCa cells have been reported to be more contractile than healthy cells, how cell contractility changes with increasing PCa metastatic potential has remained unknown. Here, we characterize the biophysical changes of PCa cells of various metastatic potential as a function of microenvironment stiffness. Using a panel of progressively increasing metastatic potential cell lines (22RV1, LNCaP, DU145, and PC3), we quantified their contractility using traction force microscopy (TFM), and measured their cortical stiffness using optical magnetic twisting cytometry (OMTC) and their motility using time-lapse microscopy. We found that PCa contractility, cell stiffness, and motility do not universally scale with metastatic potential. Rather, PCa cells of various metastatic efficiencies exhibit unique biophysical responses that are differentially influenced by substrate stiffness. Despite this biophysical diversity, this work concludes that mechanical microenvironment is a key determinant in the biophysical response of PCa with variable metastatic potentials. The mechanics-oriented focus and methodology of the study is unique and complementary to conventional biochemical and genetic strategies typically used to understand this disease, and thus may usher in new perspectives and approaches.
Collapse
Affiliation(s)
- Clayton W. Molter
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Eliana F. Muszynski
- Department of Bioengineering, McGill University, Montreal, QC, Canada
- Department of Neuroscience, McGill University, Montreal, QC, Canada
| | - Yuanyuan Tao
- Department of Bioengineering, McGill University, Montreal, QC, Canada
- Department of Electrical and Computer Engineering, McGill University, Montreal, QC, Canada
| | - Tanisha Trivedi
- Department of Bioengineering, McGill University, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Anna Clouvel
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | - Allen J. Ehrlicher
- Department of Bioengineering, McGill University, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| |
Collapse
|
15
|
Zancla A, Mozetic P, Orsini M, Forte G, Rainer A. A primer to traction force microscopy. J Biol Chem 2022; 298:101867. [PMID: 35351517 PMCID: PMC9092999 DOI: 10.1016/j.jbc.2022.101867] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 12/24/2022] Open
Abstract
Traction force microscopy (TFM) has emerged as a versatile technique for the measurement of single-cell-generated forces. TFM has gained wide use among mechanobiology laboratories, and several variants of the original methodology have been proposed. However, issues related to the experimental setup and, most importantly, data analysis of cell traction datasets may restrain the adoption of TFM by a wider community. In this review, we summarize the state of the art in TFM-related research, with a focus on the analytical methods underlying data analysis. We aim to provide the reader with a friendly compendium underlying the potential of TFM and emphasizing the methodological framework required for a thorough understanding of experimental data. We also compile a list of data analytics tools freely available to the scientific community for the furtherance of knowledge on this powerful technique.
Collapse
Affiliation(s)
- Andrea Zancla
- Department of Engineering, Università degli Studi Roma Tre, Rome, Italy; Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Pamela Mozetic
- Institute of Nanotechnology (NANOTEC), National Research Council, Lecce, Italy; Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | - Monica Orsini
- Department of Engineering, Università degli Studi Roma Tre, Rome, Italy
| | - Giancarlo Forte
- Center for Translational Medicine (CTM), International Clinical Research Center (ICRC), St Anne's University Hospital, Brno, Czechia.
| | - Alberto Rainer
- Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy; Institute of Nanotechnology (NANOTEC), National Research Council, Lecce, Italy.
| |
Collapse
|
16
|
Tsuzuki T, Baassiri K, Mahmoudi Z, Perumal AS, Rajendran K, Rubies GM, Nicolau DV. Hydrophobic Recovery of PDMS Surfaces in Contact with Hydrophilic Entities: Relevance to Biomedical Devices. MATERIALS (BASEL, SWITZERLAND) 2022; 15:2313. [PMID: 35329765 PMCID: PMC8950181 DOI: 10.3390/ma15062313] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/08/2022] [Accepted: 03/16/2022] [Indexed: 11/30/2022]
Abstract
Polydimethylsiloxane (PDMS), a silicone elastomer, is increasingly being used in health and biomedical fields due to its excellent optical and mechanical properties. Its biocompatibility and resistance to biodegradation led to various applications (e.g., lung on a chip replicating blood flow, medical interventions, and diagnostics). The many advantages of PDMS are, however, partially offset by its inherent hydrophobicity, which makes it unsuitable for applications needing wetting, thus requiring the hydrophilization of its surface by exposure to UV or O2 plasma. Yet, the elastomeric state of PDMS translates in a slow, hours to days, process of reducing its surface hydrophilicity-a process denominated as hydrophobic recovery. Using Fourier transform infrared spectroscopy (FTIR) and atomic force microscopy (AFM), the present study details the dynamics of hydrophobic recovery of PDMS, on flat bare surfaces and on surfaces embedded with hydrophilic beads. It was found that a thin, stiff, hydrophilic, silica film formed on top of the PDMS material, following its hydrophilization by UV radiation. The hydrophobic recovery of bare PDMS material is the result of an overlap of various nano-mechanical, and diffusional processes, each with its own dynamics rate, which were analyzed in parallel. The hydrophobic recovery presents a hysteresis, with surface hydrophobicity recovering only partially due to a thin, but resilient top silica layer. The monitoring of hydrophobic recovery of PDMS embedded with hydrophilic beads revealed that this is delayed, and then totally stalled in the few-micrometer vicinity of the embedded hydrophilic beads. This region where the hydrophobic recovery stalls can be used as a good approximation of the depth of the resilient, moderately hydrophilic top layer on the PDMS material. The complex processes of hydrophilization and subsequent hydrophobic recovery impact the design, fabrication, and operation of PDMS materials and devices used for diagnostics and medical procedures. Consequently, especially considering the emergence of new surgical procedures using elastomers, the impact of hydrophobic recovery on the surface of PDMS warrants more comprehensive studies.
Collapse
Affiliation(s)
- Tomoo Tsuzuki
- Faculty of Engineering and Industrial Science, Industrial Research Institute Swinburne, Swinburne University of Technology, Melbourne, VIC 3122, Australia;
| | - Karine Baassiri
- Department of Bioengineering, Faculty of Engineering, McGill University, Montreal, QC H3A 0E9, Canada; (K.B.); (Z.M.); (A.S.P.); (K.R.); (G.M.R.)
| | - Zahra Mahmoudi
- Department of Bioengineering, Faculty of Engineering, McGill University, Montreal, QC H3A 0E9, Canada; (K.B.); (Z.M.); (A.S.P.); (K.R.); (G.M.R.)
| | - Ayyappasamy Sudalaiyadum Perumal
- Department of Bioengineering, Faculty of Engineering, McGill University, Montreal, QC H3A 0E9, Canada; (K.B.); (Z.M.); (A.S.P.); (K.R.); (G.M.R.)
| | - Kavya Rajendran
- Department of Bioengineering, Faculty of Engineering, McGill University, Montreal, QC H3A 0E9, Canada; (K.B.); (Z.M.); (A.S.P.); (K.R.); (G.M.R.)
| | - Gala Montiel Rubies
- Department of Bioengineering, Faculty of Engineering, McGill University, Montreal, QC H3A 0E9, Canada; (K.B.); (Z.M.); (A.S.P.); (K.R.); (G.M.R.)
| | - Dan V. Nicolau
- Faculty of Engineering and Industrial Science, Industrial Research Institute Swinburne, Swinburne University of Technology, Melbourne, VIC 3122, Australia;
- Department of Bioengineering, Faculty of Engineering, McGill University, Montreal, QC H3A 0E9, Canada; (K.B.); (Z.M.); (A.S.P.); (K.R.); (G.M.R.)
| |
Collapse
|
17
|
Sutton AA, Molter CW, Amini A, Idicula J, Furman M, Tirgar P, Tao Y, Ghagre A, Koushki N, Khavari A, Ehrlicher AJ. Cell monolayer deformation microscopy reveals mechanical fragility of cell monolayers following EMT. Biophys J 2022; 121:629-643. [PMID: 34999131 PMCID: PMC8873957 DOI: 10.1016/j.bpj.2022.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/26/2021] [Accepted: 01/05/2022] [Indexed: 11/24/2022] Open
Abstract
Tissue and cell mechanics are crucial factors in maintaining homeostasis and in development, with aberrant mechanics contributing to many diseases. During the epithelial-to-mesenchymal transition (EMT), a highly conserved cellular program in organismal development and cancer metastasis, cells gain the ability to detach from their original location and autonomously migrate. While a great deal of biochemical and biophysical changes at the single-cell level have been revealed, how the physical properties of multicellular assemblies change during EMT, and how this may affect disease progression, is unknown. Here we introduce cell monolayer deformation microscopy (CMDM), a new methodology to measure the planar mechanical properties of cell monolayers by locally applying strain and measuring their resistance to deformation. We employ this new method to characterize epithelial multicellular mechanics at early and late stages of EMT, finding the epithelial monolayers to be relatively compliant, ductile, and mechanically homogeneous. By comparison, the transformed mesenchymal monolayers, while much stiffer, were also more brittle, mechanically heterogeneous, displayed more viscoelastic creep, and showed sharp yield points at significantly lower strains. Here, CMDM measurements identify specific biophysical functional states of EMT and offer insight into how cell aggregates fragment under mechanical stress. This mechanical fingerprinting of multicellular assemblies using new quantitative metrics may also offer new diagnostic applications in healthcare to characterize multicellular mechanical changes in disease.
Collapse
Affiliation(s)
- Amy A. Sutton
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Clayton W. Molter
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Ali Amini
- Department of Mechanical Engineering, McGill University, Montreal, Quebec, Canada
| | - Johanan Idicula
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Max Furman
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Pouria Tirgar
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Yuanyuan Tao
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Ajinkya Ghagre
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Newsha Koushki
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Adele Khavari
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Allen J. Ehrlicher
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada,Department of Mechanical Engineering, McGill University, Montreal, Quebec, Canada,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada,Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada,Centre for Structural Biology, McGill University, Montreal, Quebec, Canada,Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada,Corresponding author
| |
Collapse
|
18
|
Ramis J, Middlewick R, Pappalardo F, Cairns JT, Stewart ID, John AE, Naveed SUN, Krishnan R, Miller S, Shaw DE, Brightling CE, Buttery L, Rose F, Jenkins G, Johnson SR, Tatler AL. Lysyl oxidase-like 2 is increased in asthma and contributes to asthmatic airway remodelling. Eur Respir J 2022; 60:13993003.04361-2020. [PMID: 34996828 PMCID: PMC9260127 DOI: 10.1183/13993003.04361-2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 11/08/2021] [Indexed: 12/04/2022]
Abstract
Background Airway smooth muscle (ASM) cells are fundamental to asthma pathogenesis, influencing bronchoconstriction, airway hyperresponsiveness and airway remodelling. The extracellular matrix (ECM) can influence tissue remodelling pathways; however, to date no study has investigated the effect of ASM ECM stiffness and cross-linking on the development of asthmatic airway remodelling. We hypothesised that transforming growth factor-β (TGF-β) activation by ASM cells is influenced by ECM in asthma and sought to investigate the mechanisms involved. Methods This study combines in vitro and in vivo approaches: human ASM cells were used in vitro to investigate basal TGF-β activation and expression of ECM cross-linking enzymes. Human bronchial biopsies from asthmatic and nonasthmatic donors were used to confirm lysyl oxidase like 2 (LOXL2) expression in ASM. A chronic ovalbumin (OVA) model of asthma was used to study the effect of LOXL2 inhibition on airway remodelling. Results We found that asthmatic ASM cells activated more TGF-β basally than nonasthmatic controls and that diseased cell-derived ECM influences levels of TGF-β activated. Our data demonstrate that the ECM cross-linking enzyme LOXL2 is increased in asthmatic ASM cells and in bronchial biopsies. Crucially, we show that LOXL2 inhibition reduces ECM stiffness and TGF-β activation in vitro, and can reduce subepithelial collagen deposition and ASM thickness, two features of airway remodelling, in an OVA mouse model of asthma. Conclusion These data are the first to highlight a role for LOXL2 in the development of asthmatic airway remodelling and suggest that LOXL2 inhibition warrants further investigation as a potential therapy to reduce remodelling of the airways in severe asthma. Novel role for matrix cross-linking enzyme LOXL2 in asthmatic airway remodelling: LOXL2 is increased in #asthma but LOXL2 inhibition reduces matrix stiffness in airway smooth muscle cells and reduces remodelling in vivohttps://bit.ly/3FnzGb3
Collapse
Affiliation(s)
- Jopeth Ramis
- Biodiscovery Institute, University of Nottingham, UK.,Department of Chemical Engineering, Technological Institute of the Philippines, Philippines
| | - Robert Middlewick
- Centre for Respiratory Research/ NIHR Biomedical Research Centre, School of Medicine, University of Nottingham, UK
| | | | - Jennifer T Cairns
- Centre for Respiratory Research/ NIHR Biomedical Research Centre, School of Medicine, University of Nottingham, UK
| | - Iain D Stewart
- Centre for Respiratory Research/ NIHR Biomedical Research Centre, School of Medicine, University of Nottingham, UK.,Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, UK
| | - Alison E John
- Centre for Respiratory Research/ NIHR Biomedical Research Centre, School of Medicine, University of Nottingham, UK.,Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, UK
| | - Shams-Un-Nisa Naveed
- Centre for Respiratory Research/ NIHR Biomedical Research Centre, School of Medicine, University of Nottingham, UK.,Institute for Lung Health, Leicester NIHR Biomedical Research Centre, University of Leicester, UK
| | - Ramaswamy Krishnan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, USA
| | - Suzanne Miller
- Biodiscovery Institute, University of Nottingham, UK.,Centre for Respiratory Research/ NIHR Biomedical Research Centre, School of Medicine, University of Nottingham, UK
| | - Dominick E Shaw
- Centre for Respiratory Research/ NIHR Biomedical Research Centre, School of Medicine, University of Nottingham, UK
| | - Christopher E Brightling
- Institute for Lung Health, Leicester NIHR Biomedical Research Centre, University of Leicester, UK
| | - Lee Buttery
- Biodiscovery Institute, University of Nottingham, UK
| | - Felicity Rose
- Biodiscovery Institute, University of Nottingham, UK
| | - Gisli Jenkins
- Centre for Respiratory Research/ NIHR Biomedical Research Centre, School of Medicine, University of Nottingham, UK.,Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, UK
| | - Simon R Johnson
- Biodiscovery Institute, University of Nottingham, UK.,Centre for Respiratory Research/ NIHR Biomedical Research Centre, School of Medicine, University of Nottingham, UK
| | - Amanda L Tatler
- Centre for Respiratory Research/ NIHR Biomedical Research Centre, School of Medicine, University of Nottingham, UK
| |
Collapse
|
19
|
Substrate stiffening promotes VEGF-A functions via the PI3K/Akt/mTOR pathway. Biochem Biophys Res Commun 2022; 586:27-33. [PMID: 34823219 PMCID: PMC8785232 DOI: 10.1016/j.bbrc.2021.11.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 01/03/2023]
Abstract
While it is now well-established that substrate stiffness regulates vascular endothelial growth factor-A (VEGF-A) mediated signaling and functions, causal mechanisms remain poorly understood. Here, we report an underlying role for the PI3K/Akt/mTOR signaling pathway. This pathway is activated on stiffer substrates, is amplified by VEGF-A stimulation, and correlates with enhanced endothelial cell (EC) proliferation, contraction, pro-angiogenic secretion, and capillary-like tube formation. In the settings of advanced age-related macular degeneration, characterized by EC and retinal pigment epithelial (RPE)-mediated angiogenesis, these data implicate substrate stiffness as a novel causative mechanism and Akt/mTOR inhibition as a novel therapeutic pathway.
Collapse
|
20
|
Guo J, Jiang H, Oguntuyo K, Rios B, Boodram Z, Huebsch N. Interplay of Genotype and Substrate Stiffness in Driving the Hypertrophic Cardiomyopathy Phenotype in iPSC-Micro-Heart Muscle Arrays. Cell Mol Bioeng 2021; 14:409-425. [PMID: 34777601 PMCID: PMC8548480 DOI: 10.1007/s12195-021-00684-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/04/2021] [Indexed: 10/21/2022] Open
Abstract
INTRODUCTION In clinical and animal studies, Hypertrophic Cardiomyopathy (HCM) shares many similarities with non-inherited cardiac hypertrophy induced by pressure overload (hypertension). This suggests a potential role for mechanical stress in priming tissues with mutation-induced changes in the sarcomere to develop phenotypes associated with HCM, including hypercontractility and aberrant calcium handling. Here, we tested the hypothesis that heterozygous loss of function of Myosin Binding Protein C (MYBCP3 +/- , mutations in which account for almost 50% of inherited HCM) combines with environmental stiffness to drive HCM phenotypes. METHODS We differentiated isogenic control (WTC) and MYBPC3 +/- iPSC into cardiomyocytes using small molecule manipulation of Wnt signaling, and then purified them using lactate media. The purified cardiomyocytes were seeded into "dog bone" shaped stencil molds to form micro-heart muscle arrays (μHM). To mimic changes in myocardial stiffness stemming from pressure overload, we varied the rigidity of the substrates μHM contract against. Stiffness levels ranged from those corresponding to fetal (5 kPa), healthy (15 kPa), pre-fibrotic (30 kPa) to fibrotic (65 kPa) myocardium. Substrates were embedded with a thin layer of fluorescent beads to track contractile force, and parent iPSC were engineered to express the genetic calcium indicator, GCaMP6f. High speed video microscopy and image analysis were used to quantify calcium handling and contractility of μHM. RESULTS Substrate rigidity triggered physiological adaptation for both genotypes. However, MYBPC3 +/- μHM showed a lower tolerance to substrate stiffness with the peak traction on 15 kPa, while WTC μHM had peak traction on 30 kPa. MYBPC3 +/- μHM exhibited hypercontractility, which was exaggerated by substrate rigidity. MYBPC3 +/- μHM hypercontractility was associated with longer rise times for calcium uptake and force development, along with higher overall Ca2+ intake. CONCLUSION We found MYBPC3 +/- mutations cause iPSC-μHM to exhibit hypercontractility, and also a lower tolerance for mechanical stiffness. Understanding how genetics work in combination with mechanical stiffness to trigger and/or exacerbate pathophysiology may lead to more effective therapies for HCM. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at (10.1007/s12195-021-00684-x).
Collapse
Affiliation(s)
- Jingxuan Guo
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, Saint Louis, USA
| | - Huanzhu Jiang
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
| | - Kasoorelope Oguntuyo
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
| | - Brandon Rios
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
| | - Zoë Boodram
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
- NSF Science and Technology Center for Engineering Mechanobiology, McKelvey School of Engineering, Saint Louis, USA
- Center for Cardiovascular Research, Center for Regenerative Medicine, Center for Investigation of Membrane Excitability Diseases, Washington University in Saint Louis, Saint Louis, USA
| |
Collapse
|
21
|
Nayak AP, Lim JM, Arbel E, Wang R, Villalba DR, Nguyen TL, Schaible N, Krishnan R, Tang DD, Penn RB. Cooperativity between β-agonists and c-Abl inhibitors in regulating airway smooth muscle relaxation. FASEB J 2021; 35:e21674. [PMID: 34115899 DOI: 10.1096/fj.202100154r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 01/29/2023]
Abstract
Current therapeutic approaches to avoid or reverse bronchoconstriction rely primarily on β2 adrenoceptor agonists (β-agonists) that regulate pharmacomechanical coupling/cross bridge cycling in airway smooth muscle (ASM). Targeting actin cytoskeleton polymerization in ASM represents an alternative means to regulate ASM contraction. Herein we report the cooperative effects of targeting these distinct pathways with β-agonists and inhibitors of the mammalian Abelson tyrosine kinase (Abl1 or c-Abl). The cooperative effect of β-agonists (isoproterenol) and c-Abl inhibitors (GNF-5, or imatinib) on contractile agonist (methacholine, or histamine) -induced ASM contraction was assessed in cultured human ASM cells (using Fourier Transfer Traction Microscopy), in murine precision cut lung slices, and in vivo (flexiVent in mice). Regulation of intracellular signaling that regulates contraction (pMLC20, pMYPT1, pHSP20), and actin polymerization state (F:G actin ratio) were assessed in cultured primary human ASM cells. In each (cell, tissue, in vivo) model, c-Abl inhibitors and β-agonist exhibited additive effects in either preventing or reversing ASM contraction. Treatment of contracted ASM cells with c-Abl inhibitors and β-agonist cooperatively increased actin disassembly as evidenced by a significant reduction in the F:G actin ratio. Mechanistic studies indicated that the inhibition of pharmacomechanical coupling by β-agonists is near optimal and is not increased by c-Abl inhibitors, and the cooperative effect on ASM relaxation resides in further relaxation of ASM tension development caused by actin cytoskeleton depolymerization, which is regulated by both β-agonists and c-Abl inhibitors. Thus, targeting actin cytoskeleton polymerization represents an untapped therapeutic reserve for managing airway resistance.
Collapse
Affiliation(s)
- Ajay P Nayak
- Department of Medicine, Pulmonary and Critical Care Medicine, Center for Translational Medicine, Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, PA, USA
| | - John M Lim
- Department of Medicine, Pulmonary and Critical Care Medicine, Center for Translational Medicine, Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, PA, USA
| | - Eylon Arbel
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Ruping Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Dominic R Villalba
- Department of Medicine, Pulmonary and Critical Care Medicine, Center for Translational Medicine, Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, PA, USA
| | - Tahn L Nguyen
- Department of Medicine, Pulmonary and Critical Care Medicine, Center for Translational Medicine, Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, PA, USA
| | - Niccole Schaible
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ramaswamy Krishnan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Dale D Tang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Raymond B Penn
- Department of Medicine, Pulmonary and Critical Care Medicine, Center for Translational Medicine, Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
22
|
Ehlinger C, Mathieu E, Rabineau M, Ball V, Lavalle P, Haikel Y, Vautier D, Kocgozlu L. Insensitivity of dental pulp stem cells migration to substrate stiffness. Biomaterials 2021; 275:120969. [PMID: 34157563 DOI: 10.1016/j.biomaterials.2021.120969] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/26/2021] [Accepted: 06/09/2021] [Indexed: 12/16/2022]
Abstract
Dental pulp stem cells (DPSCs) are a promising cell source for regeneration of dental pulp. Migration is a key event but influence of the microenvironment rigidity (5 kPa at the center of dental pulp to 20 GPa for the dentin) is largely unknown. Mechanical signals are transmitted from the extracellular matrix to the cytoskeleton, to the nuclei, and to the chromatin, potentially regulating gene expression. To identify the microenvironmental influence on migration, we analyzed motility on PDMS substrates with stiffness increasing from 1.5 kPa up to 2.5 MPa. We found that migration speed slightly increases as substrate stiffness decreases in correlation with decreasing focal adhesion size. Motility is relatively insensitive to substrate stiffness, even on a bi-rigidity PDMS substrate where DPSCs migrate without preferential direction. Migration is independent of both myosin II activity and YAP translocation after myosin II inhibition. Additionally, inhibition of Arp2/3 complex leads to significant speed decrease for all rigidities, suggesting contribution of the lamellipodia in the migration. Interestingly, the chromatin architecture remains stable after a 7-days exposure on the PDMS substrates for all rigidity. To design scaffold mimicking dental pulp environment, similar DPSCs migration for all rigidity, leaves field open to choose this mechanical parameter.
Collapse
Affiliation(s)
- Claire Ehlinger
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France
| | - Eric Mathieu
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France
| | - Morgane Rabineau
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France
| | - Vincent Ball
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France
| | - Philippe Lavalle
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France
| | - Youssef Haikel
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France
| | - Dominique Vautier
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France.
| | - Leyla Kocgozlu
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France.
| |
Collapse
|
23
|
Ghagre A, Amini A, Srivastava LK, Tirgar P, Khavari A, Koushki N, Ehrlicher A. Pattern-Based Contractility Screening, a Reference-Free Alternative to Traction Force Microscopy Methodology. ACS APPLIED MATERIALS & INTERFACES 2021; 13:19726-19735. [PMID: 33884863 DOI: 10.1021/acsami.1c02987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The sensing and generation of cellular forces are essential aspects of life. Traction force microscopy (TFM) has emerged as a standard broadly applicable methodology to measure cell contractility and its role in cell behavior. While TFM platforms have enabled diverse discoveries, their implementation remains limited in part due to various constraints, such as time-consuming substrate fabrication techniques, the need to detach cells to measure null force images, followed by complex imaging and analysis, and the unavailability of cells for postprocessing. Here we introduce a reference-free technique to measure cell contractile work in real time, with commonly available substrate fabrication methodologies, simple imaging, and analysis with the availability of the cells for postprocessing. In this technique, we confine the cells on fluorescent adhesive protein micropatterns of a known area on compliant silicone substrates and use the cell deformed pattern area to calculate cell contractile work. We validated this approach by comparing this pattern-based contractility screening (PaCS) with conventional bead-displacement TFM and show quantitative agreement between the methodologies. Using this platform, we measure the contractile work of highly metastatic MDA-MB-231 breast cancer cells that is significantly higher than the contractile work of noninvasive MCF-7 cells. PaCS enables the broader implementation of contractile work measurements in diverse quantitative biology and biomedical applications.
Collapse
Affiliation(s)
- Ajinkya Ghagre
- Department of Bioengineering, McGill University, Montreal H3A 0E9, Canada
| | - Ali Amini
- Department of Mechanical Engineering, McGill University, Montreal H3A 0C3, Canada
| | | | - Pouria Tirgar
- Department of Bioengineering, McGill University, Montreal H3A 0E9, Canada
| | - Adele Khavari
- Department of Bioengineering, McGill University, Montreal H3A 0E9, Canada
| | - Newsha Koushki
- Department of Bioengineering, McGill University, Montreal H3A 0E9, Canada
| | - Allen Ehrlicher
- Department of Bioengineering, McGill University, Montreal H3A 0E9, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal H3A 0C7, Canada
- Department of Mechanical Engineering, McGill University, Montreal H3A 0C3, Canada
- Department of Biomedical Engineering, McGill University, Montreal H3A 2B4, Quebec, Canada
- Centre for Structural Biology, McGill University, Montreal H3A 0G4, Quebec, Canada
- Goodman Cancer Research Centre, McGill University, Montreal H3A 1A3, Quebec, Canada
| |
Collapse
|
24
|
Rheinlaender J, Wirbel H, Schäffer TE. Spatial correlation of cell stiffness and traction forces in cancer cells measured with combined SICM and TFM. RSC Adv 2021; 11:13951-13956. [PMID: 35423943 PMCID: PMC8697701 DOI: 10.1039/d1ra01277k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/06/2021] [Indexed: 12/31/2022] Open
Abstract
The mechanical properties of cancer cells at the single-cell and the subcellular level might be the key for answering long-standing questions in the diagnosis and treatment of cancer. However, the subcellular distribution of two main mechanical properties, cell stiffness and traction forces, has been investigated only rarely and qualitatively yet. Here, we present the first direct combination of scanning ion conductance microscopy (SICM) and traction force microscopy (TFM), which we used to identify a correlation between the local stiffness and the local traction force density in living cells. We found a correlation in normal breast epithelial cells, but no correlation in cancerous breast epithelial cells. This indicates that the interplay between cell stiffness and traction forces is altered in cancer cells as compared to healthy cells, which might give new insight in the research field of cancer cell mechanobiology.
Collapse
Affiliation(s)
- Johannes Rheinlaender
- Institute of Applied Physics, University of Tübingen Auf der Morgenstelle 10 72076 Tübingen Germany +49 7071 29 5093 +49 7071 29 76030
| | - Hannes Wirbel
- Institute of Applied Physics, University of Tübingen Auf der Morgenstelle 10 72076 Tübingen Germany +49 7071 29 5093 +49 7071 29 76030
| | - Tilman E Schäffer
- Institute of Applied Physics, University of Tübingen Auf der Morgenstelle 10 72076 Tübingen Germany +49 7071 29 5093 +49 7071 29 76030
| |
Collapse
|
25
|
Hnatiuk AP, Briganti F, Staudt DW, Mercola M. Human iPSC modeling of heart disease for drug development. Cell Chem Biol 2021; 28:271-282. [PMID: 33740432 PMCID: PMC8054828 DOI: 10.1016/j.chembiol.2021.02.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/26/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) have emerged as a promising platform for pharmacogenomics and drug development. In cardiology, they make it possible to produce unlimited numbers of patient-specific human cells that reproduce hallmark features of heart disease in the culture dish. Their potential applications include the discovery of mechanism-specific therapeutics, the evaluation of safety and efficacy in a human context before a drug candidate reaches patients, and the stratification of patients for clinical trials. Although this new technology has the potential to revolutionize drug discovery, translational hurdles have hindered its widespread adoption for pharmaceutical development. Here we discuss recent progress in overcoming these hurdles that should facilitate the use of hiPSCs to develop new medicines and individualize therapies for heart disease.
Collapse
Affiliation(s)
- Anna P Hnatiuk
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Francesca Briganti
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - David W Staudt
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Mark Mercola
- Stanford Cardiovascular Institute, 240 Pasteur Drive, Biomedical Innovation Building, Palo Alto, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
26
|
Acheva A, Kärki T, Schaible N, Krishnan R, Tojkander S. Adipokine Leptin Co-operates With Mechanosensitive Ca 2 +-Channels and Triggers Actomyosin-Mediated Motility of Breast Epithelial Cells. Front Cell Dev Biol 2021; 8:607038. [PMID: 33490070 PMCID: PMC7815691 DOI: 10.3389/fcell.2020.607038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022] Open
Abstract
In postmenopausal women, a major risk factor for the development of breast cancer is obesity. In particular, the adipose tissue-derived adipokine leptin has been strongly linked to tumor cell proliferation, migration, and metastasis, but the underlying mechanisms remain unclear. Here we show that treatment of normal mammary epithelial cells with leptin induces EMT-like features characterized by higher cellular migration speeds, loss of structural ordering of 3D-mammo spheres, and enhancement of epithelial traction forces. Mechanistically, leptin triggers the phosphorylation of myosin light chain kinase-2 (MLC-2) through the interdependent activity of leptin receptor and Ca2+ channels. These data provide evidence that leptin-activated leptin receptors, in co-operation with mechanosensitive Ca2+ channels, play a role in the development of breast carcinomas through the regulation of actomyosin dynamics.
Collapse
Affiliation(s)
- Anna Acheva
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Tytti Kärki
- Department of Applied Physics, School of Science, Aalto University, Espoo, Finland
| | - Niccole Schaible
- Beth Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Ramaswamy Krishnan
- Beth Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Sari Tojkander
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
27
|
Yang S, Valencia FR, Sabass B, Plotnikov SV. Quantitative Analysis of Myofibroblast Contraction by Traction Force Microscopy. Methods Mol Biol 2021; 2299:181-195. [PMID: 34028744 DOI: 10.1007/978-1-0716-1382-5_14] [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] [Indexed: 06/12/2023]
Abstract
Myofibroblasts play important roles in physiological processes such as wound healing and tissue repair. While high contractile forces generated by the actomyosin network enable myofibroblasts to physically contract the wound and bring together injured tissue, prolonged and elevated levels of contraction also drive the progression of fibrosis and cancer. However, quantitative mapping of these forces has been difficult due to their extremely low magnitude ranging from 100 pN/μm2 to 2 nN/μm2. Here, we provide a protocol to measure cellular forces exerted on two-dimensional compliant elastic hydrogels. We describe the fabrication of polyacrylamide hydrogels labeled with fluorescent fiducial markers, functionalization of substrates with ECM proteins, setting up the experiment, and imaging procedures. We demonstrate the application of this technique for quantitative analysis of traction forces exerted by myofibroblasts.
Collapse
Affiliation(s)
- Shuying Yang
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Fernando R Valencia
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Benedikt Sabass
- Theoretical Physics of Living Matter, Institute of Biological Information Processing and Institute of Advanced Simulation, Forschungszentrum Juelich, Juelich, Germany
- Department of Veterinary Sciences, Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sergey V Plotnikov
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
28
|
Missirlis D, Haraszti T, Heckmann L, Spatz JP. Substrate Resistance to Traction Forces Controls Fibroblast Polarization. Biophys J 2020; 119:2558-2572. [PMID: 33217384 DOI: 10.1016/j.bpj.2020.10.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/25/2022] Open
Abstract
The mechanics of fibronectin-rich extracellular matrix regulate cell physiology in a number of diseases, prompting efforts to elucidate cell mechanosensing mechanisms at the molecular and cellular scale. Here, the use of fibronectin-functionalized silicone elastomers that exhibit considerable frequency dependence in viscoelastic properties unveiled the presence of two cellular processes that respond discreetly to substrate mechanical properties. Weakly cross-linked elastomers supported efficient focal adhesion maturation and fibroblast spreading because of an apparent stiff surface layer. However, they did not enable cytoskeletal and fibroblast polarization; elastomers with high cross-linking and low deformability were required for polarization. Our results suggest as an underlying reason for this behavior the inability of soft elastomer substrates to resist traction forces rather than a lack of sufficient traction force generation. Accordingly, mild inhibition of actomyosin contractility rescued fibroblast polarization even on the softer elastomers. Our findings demonstrate differential dependence of substrate physical properties on distinct mechanosensitive processes and provide a premise to reconcile previously proposed local and global models of cell mechanosensing.
Collapse
Affiliation(s)
- Dimitris Missirlis
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Heidelberg, Germany.
| | - Tamás Haraszti
- DWI-Leibniz Institute for Interactive Materials, Aachen, Germany; RWTH Aachen University, Institute for Technical and Macromolecular Chemistry, Aachen, Germany
| | - Lara Heckmann
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Heidelberg, Germany
| | - Joachim P Spatz
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Heidelberg, Germany; Heidelberg University, Department of Biophysical Chemistry, Physical Chemistry Institute, Heidelberg, Germany
| |
Collapse
|
29
|
Guo J, Simmons DW, Ramahdita G, Munsell MK, Oguntuyo K, Kandalaft B, Rios B, Pear M, Schuftan D, Jiang H, Lake SP, Genin GM, Huebsch N. Elastomer-Grafted iPSC-Derived Micro Heart Muscles to Investigate Effects of Mechanical Loading on Physiology. ACS Biomater Sci Eng 2020; 7:2973-2989. [PMID: 34275296 DOI: 10.1021/acsbiomaterials.0c00318] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Mechanical loading plays a critical role in cardiac pathophysiology. Engineered heart tissues derived from human induced pluripotent stem cells (iPSCs) allow rigorous investigations of the molecular and pathophysiological consequences of mechanical cues. However, many engineered heart muscle models have complex fabrication processes and require large cell numbers, making it difficult to use them together with iPSC-derived cardiomyocytes to study the influence of mechanical loading on pharmacology and genotype-phenotype relationships. To address this challenge, simple and scalable iPSC-derived micro-heart-muscle arrays (μHM) have been developed. "Dog-bone-shaped" molds define the boundary conditions for tissue formation. Here, we extend the μHM model by forming these tissues on elastomeric substrates with stiffnesses spanning from 5 to 30 kPa. Tissue assembly was achieved by covalently grafting fibronectin to the substrate. Compared to μHM formed on plastic, elastomer-grafted μHM exhibited a similar gross morphology, sarcomere assembly, and tissue alignment. When these tissues were formed on substrates with different elasticity, we observed marked shifts in contractility. Increased contractility was correlated with increases in calcium flux and a slight increase in cell size. This afterload-enhanced μHM system enables mechanical control of μHM and real-time tissue traction force microscopy for cardiac physiology measurements, providing a dynamic tool for studying pathophysiology and pharmacology.
Collapse
Affiliation(s)
- Jingxuan Guo
- Department of Mechanical Engineering and Materials Science, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States
| | - Daniel W Simmons
- Department of Biomedical Engineering, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States.,NSF Science and Technology Center for Engineering Mechanobiology, McKelvey School of Engineering, 1 Brookings Dr., St. Louis, Missouri 63130, United States
| | - Ghiska Ramahdita
- Department of Mechanical Engineering and Materials Science, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States.,NSF Science and Technology Center for Engineering Mechanobiology, McKelvey School of Engineering, 1 Brookings Dr., St. Louis, Missouri 63130, United States
| | - Mary K Munsell
- Department of Biomedical Engineering, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States
| | - Kasoorelope Oguntuyo
- Department of Biomedical Engineering, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States
| | - Brennan Kandalaft
- Department of Biomedical Engineering, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States
| | - Brandon Rios
- Department of Biomedical Engineering, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States
| | - Missy Pear
- Department of Biomedical Engineering, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States
| | - David Schuftan
- Department of Biomedical Engineering, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States
| | - Huanzhu Jiang
- Department of Biomedical Engineering, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States
| | - Spencer P Lake
- Department of Mechanical Engineering and Materials Science, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States
| | - Guy M Genin
- Department of Mechanical Engineering and Materials Science, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States.,NSF Science and Technology Center for Engineering Mechanobiology, McKelvey School of Engineering, 1 Brookings Dr., St. Louis, Missouri 63130, United States
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States.,NSF Science and Technology Center for Engineering Mechanobiology, McKelvey School of Engineering, 1 Brookings Dr., St. Louis, Missouri 63130, United States.,Center for Cardiovascular Research, Center for Regenerative Medicine, Center for Investigation of Membrane Excitability Diseases, Washington University in Saint Louis, University City, St. Louis, Missouri 63130, United States
| |
Collapse
|
30
|
Stasiak SE, Jamieson RR, Bouffard J, Cram EJ, Parameswaran H. Intercellular communication controls agonist-induced calcium oscillations independently of gap junctions in smooth muscle cells. SCIENCE ADVANCES 2020; 6:eaba1149. [PMID: 32821820 PMCID: PMC7406377 DOI: 10.1126/sciadv.aba1149] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 06/18/2020] [Indexed: 06/11/2023]
Abstract
In this study, we report the existence of a communication system among human smooth muscle cells that uses mechanical forces to frequency modulate long-range calcium waves. An important consequence of this mechanical signaling is that changes in stiffness of the underlying extracellular matrix can interfere with the frequency modulation of Ca2+ waves, causing smooth muscle cells from healthy human donors to falsely perceive a much higher agonist dose than they actually received. This aberrant sensing of contractile agonist dose on stiffer matrices is completely absent in isolated smooth muscle cells, although the isolated cells can sense matrix rigidity. We show that the intercellular communication that enables this collective Ca2+ response in smooth muscle cells does not involve transport across gap junctions or extracellular diffusion of signaling molecules. Instead, our data support a collective model in which mechanical signaling among smooth muscle cells regulates their response to contractile agonists.
Collapse
Affiliation(s)
- S. E. Stasiak
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - R. R. Jamieson
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - J. Bouffard
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - E. J. Cram
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - H. Parameswaran
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
31
|
Aspergillus fumigatus Protease Alkaline Protease 1 (Alp1): A New Therapeutic Target for Fungal Asthma. J Fungi (Basel) 2020; 6:jof6020088. [PMID: 32560087 PMCID: PMC7345148 DOI: 10.3390/jof6020088] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/10/2020] [Accepted: 06/12/2020] [Indexed: 12/26/2022] Open
Abstract
We review three recent findings that have fundamentally altered our understanding of causative mechanisms underlying fungal-related asthma. These mechanisms may be partially independent of host inflammatory processes but are strongly dependent upon the actions of Alp1 on lung structural cells. They entail (i) bronchial epithelial sensing of Alp1; (ii) Alp1-induced airway smooth muscle (ASM) contraction; (iii) Alp1-induced airflow obstruction. Collectively, these mechanisms point to Alp1 as a new target for intervention in fungal asthma.
Collapse
|
32
|
Lu RA, Zeki AA, Ram-Mohan S, Nguyen N, Bai Y, Chmiel K, Pecic S, Ai X, Krishnan R, Ghosh CC. Inhibiting Airway Smooth Muscle Contraction Using Pitavastatin: A Role for the Mevalonate Pathway in Regulating Cytoskeletal Proteins. Front Pharmacol 2020; 11:469. [PMID: 32435188 PMCID: PMC7218099 DOI: 10.3389/fphar.2020.00469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/25/2020] [Indexed: 12/16/2022] Open
Abstract
Despite maximal use of currently available therapies, a significant number of asthma patients continue to experience severe, and sometimes life-threatening bronchoconstriction. To fill this therapeutic gap, we examined a potential role for the 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) inhibitor, pitavastatin. Using human airway smooth muscle (ASM) cells and murine precision-cut lung slices, we discovered that pitavastatin significantly inhibited basal-, histamine-, and methacholine (MCh)-induced ASM contraction. This occurred via reduction of myosin light chain 2 (MLC2) phosphorylation, and F-actin stress fiber density and distribution, in a mevalonate (MA)- and geranylgeranyl pyrophosphate (GGPP)-dependent manner. Pitavastatin also potentiated the ASM relaxing effect of a simulated deep breath, a beneficial effect that is notably absent with the β2-agonist, isoproterenol. Finally, pitavastatin attenuated ASM pro-inflammatory cytokine production in a GGPP-dependent manner. By targeting all three hallmark features of ASM dysfunction in asthma—contraction, failure to adequately relax in response to a deep breath, and inflammation—pitavastatin may represent a unique asthma therapeutic.
Collapse
Affiliation(s)
- Robin A Lu
- Department of Emergency Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Amir A Zeki
- Division of Pulmonary, Critical Care, and Sleep Medicine, U.C. Davis Lung Center, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Sumati Ram-Mohan
- Department of Emergency Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Nhan Nguyen
- Department of Emergency Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Yan Bai
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kenneth Chmiel
- Division of Pulmonary, Critical Care, and Sleep Medicine, U.C. Davis Lung Center, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University, Fullerton, CA, United States
| | - Xingbin Ai
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Ramaswamy Krishnan
- Department of Emergency Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Chandra C Ghosh
- Department of Emergency Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
33
|
Stubb A, Laine RF, Miihkinen M, Hamidi H, Guzmán C, Henriques R, Jacquemet G, Ivaska J. Fluctuation-Based Super-Resolution Traction Force Microscopy. NANO LETTERS 2020; 20:2230-2245. [PMID: 32142297 PMCID: PMC7146861 DOI: 10.1021/acs.nanolett.9b04083] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 03/02/2020] [Indexed: 05/24/2023]
Abstract
Cellular mechanics play a crucial role in tissue homeostasis and are often misregulated in disease. Traction force microscopy is one of the key methods that has enabled researchers to study fundamental aspects of mechanobiology; however, traction force microscopy is limited by poor resolution. Here, we propose a simplified protocol and imaging strategy that enhances the output of traction force microscopy by increasing i) achievable bead density and ii) the accuracy of bead tracking. Our approach relies on super-resolution microscopy, enabled by fluorescence fluctuation analysis. Our pipeline can be used on spinning-disk confocal or widefield microscopes and is compatible with available analysis software. In addition, we demonstrate that our workflow can be used to gain biologically relevant information and is suitable for fast long-term live measurement of traction forces even in light-sensitive cells. Finally, using fluctuation-based traction force microscopy, we observe that filopodia align to the force field generated by focal adhesions.
Collapse
Affiliation(s)
- Aki Stubb
- Turku
Bioscience Centre, University of Turku and
Åbo Akademi University, FI-20520 Turku, Finland
| | - Romain F. Laine
- MRC-Laboratory
for Molecular Cell Biology, University College
London, London WC1E 6BT, U.K.
- The
Francis Crick Institute, London NW1 1AT, U.K.
| | - Mitro Miihkinen
- Turku
Bioscience Centre, University of Turku and
Åbo Akademi University, FI-20520 Turku, Finland
| | - Hellyeh Hamidi
- Turku
Bioscience Centre, University of Turku and
Åbo Akademi University, FI-20520 Turku, Finland
| | - Camilo Guzmán
- Turku
Bioscience Centre, University of Turku and
Åbo Akademi University, FI-20520 Turku, Finland
| | - Ricardo Henriques
- MRC-Laboratory
for Molecular Cell Biology, University College
London, London WC1E 6BT, U.K.
- The
Francis Crick Institute, London NW1 1AT, U.K.
| | - Guillaume Jacquemet
- Turku
Bioscience Centre, University of Turku and
Åbo Akademi University, FI-20520 Turku, Finland
- Faculty
of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
| | - Johanna Ivaska
- Turku
Bioscience Centre, University of Turku and
Åbo Akademi University, FI-20520 Turku, Finland
- Department
of Biochemistry, University of Turku, FIN-20520 Turku, Finland
| |
Collapse
|
34
|
Multi-well plate cell contraction assay detects negatively correlated cellular responses to pharmacological inhibitors in contractility and migration. Biochem Biophys Res Commun 2020; 521:527-532. [DOI: 10.1016/j.bbrc.2019.10.160] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/22/2019] [Indexed: 12/28/2022]
|
35
|
Rausch M, Böhringer D, Steinmann M, Schubert DW, Schrüfer S, Mark C, Fabry B. Measurement of Skeletal Muscle Fiber Contractility with High-Speed Traction Microscopy. Biophys J 2019; 118:657-666. [PMID: 31952805 DOI: 10.1016/j.bpj.2019.12.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/11/2019] [Accepted: 12/11/2019] [Indexed: 01/07/2023] Open
Abstract
We describe a technique for simultaneous quantification of the contractile forces and cytosolic calcium dynamics of muscle fibers embedded in three-dimensional biopolymer gels under auxotonic loading conditions. We derive a scaling law for linear elastic matrices such as basement membrane extract hydrogels (Matrigel) that allows us to measure contractile force from the shape of the relaxed and contracted muscle cell and the Young's modulus of the matrix without further knowledge of the matrix deformations surrounding the cell and without performing computationally intensive inverse force reconstruction algorithms. We apply our method to isolated mouse flexor digitorum brevis (FDB) fibers that are embedded in 10 mg/mL Matrigel. Upon electrical stimulation, individual FDB fibers show twitch forces of 0.37 ± 0.15 μN and tetanic forces (100-Hz stimulation frequency) of 2.38 ± 0.71 μN, corresponding to a tension of 0.44 ± 0.25 kPa and 2.53 ± 1.17 kPa, respectively. Contractile forces of FDB fibers increase in response to caffeine and the troponin-calcium stabilizer tirasemtiv, similar to responses measured in whole muscle. From simultaneous high-speed measurements of cell length changes and cytosolic calcium concentration using confocal line scanning at a frequency of 2048 Hz, we show that twitch and tetanic force responses to electric pulses follow the low-pass filtered calcium signal. In summary, we present a technically simple high-speed method for measuring contractile forces and cytosolic calcium dynamics of single muscle fibers. We expect that our method will help to reduce preparation time, costs, and the number of sacrificed animals needed for experiments such as drug testing.
Collapse
Affiliation(s)
- Martin Rausch
- Novartis Institutes for BioMedical Research, Basel, Switzerland.
| | - David Böhringer
- Department of Physics, Institute for Polymer Materials, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Dirk W Schubert
- Department of Materials Science, Institute for Polymer Materials, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Schrüfer
- Department of Materials Science, Institute for Polymer Materials, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Mark
- Department of Physics, Institute for Polymer Materials, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Ben Fabry
- Department of Physics, Institute for Polymer Materials, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
36
|
Ram-Mohan S, Bai Y, Schaible N, Ehrlicher AJ, Cook DP, Suki B, Stoltz DA, Solway J, Ai X, Krishnan R. Tissue traction microscopy to quantify muscle contraction within precision-cut lung slices. Am J Physiol Lung Cell Mol Physiol 2019; 318:L323-L330. [PMID: 31774304 DOI: 10.1152/ajplung.00297.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In asthma, acute bronchospasm is driven by contractile forces of airway smooth muscle (ASM). These forces can be imaged in the cultured ASM cell or assessed in the muscle strip and the tracheal/bronchial ring, but in each case, the ASM is studied in isolation from the native airway milieu. Here, we introduce a novel platform called tissue traction microscopy (TTM) to measure ASM contractile force within porcine and human precision-cut lung slices (PCLS). Compared with the conventional measurements of lumen area changes in PCLS, TTM measurements of ASM force changes are 1) more sensitive to bronchoconstrictor stimuli, 2) less variable across airways, and 3) provide spatial information. Notably, within every human airway, TTM measurements revealed local regions of high ASM contraction that we call "stress hotspots". As an acute response to cyclic stretch, these hotspots promptly decreased but eventually recovered in magnitude, spatial location, and orientation, consistent with local ASM fluidization and resolidification. By enabling direct and precise measurements of ASM force, TTM should accelerate preclinical studies of airway reactivity.
Collapse
Affiliation(s)
- Sumati Ram-Mohan
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Yan Bai
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Niccole Schaible
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Allen J Ehrlicher
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada
| | - Daniel P Cook
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa
| | - Bela Suki
- Biomedical Engineering Department, Boston University, Boston, Massachusetts
| | - David A Stoltz
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa
| | - Julian Solway
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Xingbin Ai
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ramaswamy Krishnan
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
37
|
Redes JL, Basu T, Ram-Mohan S, Ghosh CC, Chan EC, Sek AC, Zhao M, Krishnan R, Rosenberg HF, Druey KM. Aspergillus fumigatus-Secreted Alkaline Protease 1 Mediates Airways Hyperresponsiveness in Severe Asthma. Immunohorizons 2019; 3:368-377. [PMID: 31603851 PMCID: PMC10985461 DOI: 10.4049/immunohorizons.1900046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 07/19/2019] [Indexed: 11/19/2022] Open
Abstract
The hallmark features of allergic asthma are type 2 (eosinophilic) inflammation and airways hyperresponsiveness (AHR). Although these features often comanifest in mouse lungs in vivo, we demonstrate in this study that the serine protease Alp1 from the ubiquitous mold and allergen, Aspergillus fumigatus, can induce AHR in mice unable to generate eosinophilic inflammation. Strikingly, Alp1 induced AHR in mice devoid of protease-activated receptor 2/F2 trypsin-like receptor 1 (PAR2/F2RL1), a receptor expressed in lung epithelium that is critical for allergic responses to protease-containing allergens. Instead, using precision-cut lung slices and human airway smooth muscle cells, we demonstrate that Alp1 directly increased contractile force. Taken together, these findings suggest that Alp1 induces bronchoconstriction through mechanisms that are largely independent of allergic inflammation and point to a new target for direct intervention of fungal-associated asthma.
Collapse
Affiliation(s)
- Jamie L Redes
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD 20892
| | - Trisha Basu
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD 20892
| | - Sumati Ram-Mohan
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Chandra C Ghosh
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Eunice C Chan
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD 20892
| | - Albert C Sek
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD 20892; and
| | - Ming Zhao
- Protein Chemistry Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Rockville, MD 20852
| | - Ramaswamy Krishnan
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Helene F Rosenberg
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD 20892; and
| | - Kirk M Druey
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
38
|
Polio SR, Stasiak SE, Jamieson RR, Balestrini JL, Krishnan R, Parameswaran H. Extracellular matrix stiffness regulates human airway smooth muscle contraction by altering the cell-cell coupling. Sci Rep 2019; 9:9564. [PMID: 31267003 PMCID: PMC6606622 DOI: 10.1038/s41598-019-45716-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 06/13/2019] [Indexed: 12/31/2022] Open
Abstract
For an airway or a blood vessel to narrow, there must be a connected path that links the smooth muscle (SM) cells with each other, and transmits forces around the organ, causing it to constrict. Currently, we know very little about the mechanisms that regulate force transmission pathways in a multicellular SM ensemble. Here, we used extracellular matrix (ECM) micropatterning to study force transmission in a two-cell ensemble of SM cells. Using the two-SM cell ensemble, we demonstrate (a) that ECM stiffness acts as a switch that regulates whether SM force is transmitted through the ECM or through cell-cell connections. (b) Fluorescent imaging for adherens junctions and focal adhesions show the progressive loss of cell-cell borders and the appearance of focal adhesions with the increase in ECM stiffness (confirming our mechanical measurements). (c) At the same ECM stiffness, we show that the presence of a cell-cell border substantially decreases the overall contractility of the SM cell ensemble. Our results demonstrate that connectivity among SM cells is a critical factor to consider in the development of diseases such as asthma and hypertension.
Collapse
Affiliation(s)
- Samuel R Polio
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Suzanne E Stasiak
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Ryan R Jamieson
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Jenna L Balestrini
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ramaswamy Krishnan
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, 02115, USA
| | | |
Collapse
|
39
|
Yoshie H, Koushki N, Molter C, Siegel PM, Krishnan R, Ehrlicher AJ. High Throughput Traction Force Microscopy Using PDMS Reveals Dose-Dependent Effects of Transforming Growth Factor-β on the Epithelial-to-Mesenchymal Transition. J Vis Exp 2019. [PMID: 31205302 DOI: 10.3791/59364] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Cellular contractility is essential in diverse aspects of biology, driving processes that range from motility and division, to tissue contraction and mechanical stability, and represents a core element of multi-cellular animal life. In adherent cells, acto-myosin contraction is seen in traction forces that cells exert on their substrate. Dysregulation of cellular contractility appears in a myriad of pathologies, making contractility a promising target in diverse diagnostic approaches using biophysics as a metric. Moreover, novel therapeutic strategies can be based on correcting the apparent malfunction of cell contractility. These applications, however, require direct quantification of these forces. We have developed silicone elastomer-based traction force microscopy (TFM) in a parallelized multi-well format. Our use of a silicone rubber, specifically polydimethylsiloxane (PDMS), rather than the commonly employed hydrogel polyacrylamide (PAA) enables us to make robust and inert substrates with indefinite shelf-lives requiring no specialized storage conditions. Unlike pillar-PDMS based approaches that have a modulus in the GPa range, the PDMS used here is very compliant, ranging from approximately 0.4 kPa to 100 kPa. We create a high-throughput platform for TFM by partitioning these large monolithic substrates spatially into biochemically independent wells, creating a multi-well platform for traction force screening that is compatible with existing multi-well systems. In this manuscript, we use this multi-well traction force system to examine the Epithelial to Mesenchymal Transition (EMT); we induce EMT in NMuMG cells by exposing them to TGF-β, and to quantify the biophysical changes during EMT. We measure the contractility as a function of concentration and duration of TGF-β exposure. Our findings here demonstrate the utility of parallelized TFM in the context of disease biophysics.
Collapse
Affiliation(s)
| | | | | | - Peter M Siegel
- Goodman Cancer Research Centre, McGill University; Department of Medicine, McGill University
| | | | - Allen J Ehrlicher
- Department of Bioengineering, McGill University; Goodman Cancer Research Centre, McGill University;
| |
Collapse
|
40
|
McCoey JM, de Gille RW, Nasr B, Tetienne JP, Hall LT, Simpson DA, Hollenberg LCL. Rapid, High-Resolution Magnetic Microscopy of Single Magnetic Microbeads. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1805159. [PMID: 30912265 DOI: 10.1002/smll.201805159] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/31/2019] [Indexed: 06/09/2023]
Abstract
Magnetic microparticles or "beads" are used in a variety of research applications from cell sorting through to optical force traction microscopy. The magnetic properties of such particles can be tailored for specific applications with the uniformity of individual beads critical to their function. However, the majority of magnetic characterization techniques quantify the magnetic properties from large bead ensembles. Developing new magnetic imaging techniques to evaluate and visualize the magnetic fields from single beads will allow detailed insight into the magnetic uniformity, anisotropy, and alignment of magnetic domains. Here, diamond-based magnetic microscopy is applied to image and characterize individual magnetic beads with varying magnetic and structural properties: ferromagnetic and superparamagnetic/paramagnetic, shell (coated with magnetic material), and solid (magnetic material dispersed in matrix). The single-bead magnetic images identify irregularities in the magnetic profiles from individual bead populations. Magnetic simulations account for the varying magnetic profiles and allow to infer the magnetization of individual beads. Additionally, this work shows that the imaging technique can be adapted to achieve illumination-free tracking of magnetic beads, opening the possibility of tracking cell movements and mechanics in photosensitive contexts.
Collapse
Affiliation(s)
- Julia M McCoey
- School of Physics, University of Melbourne, Parkville, 3010, Australia
| | - Robert W de Gille
- School of Physics, University of Melbourne, Parkville, 3010, Australia
| | - Babak Nasr
- The Department of Electrical and Electronic Engineering, The University of Melbourne, Victoria, 3010, Australia
| | | | - Liam T Hall
- School of Physics, University of Melbourne, Parkville, 3010, Australia
| | - David A Simpson
- School of Physics, University of Melbourne, Parkville, 3010, Australia
| | | |
Collapse
|
41
|
Bush J, Maruthamuthu V. In situ determination of exerted forces in magnetic pulling cytometry. AIP ADVANCES 2019; 9:035221. [PMID: 30915259 PMCID: PMC6417906 DOI: 10.1063/1.5084261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/05/2019] [Indexed: 06/09/2023]
Abstract
Localized application of exogenous forces on soft biomaterials and cells is often essential for the study of their response to external mechanical stimuli. Magnetic means of applying forces, particularly those based on permanent magnets and magnetic beads coupled to substrates or cells provide an accessible means of exerting forces of appropriate magnitude. The amount of force exerted, however, is often inferred from calibration performed ex situ, with typically similar but different magnetic beads. Here, we construct a simple magnetic tweezer by coupling a pencil-shaped stainless-steel probe to permanent neodymium magnets using a 3D printed adapter. We then demonstrate the in situ determination of magnetic bead pulling forces on a super-paramagnetic micro-bead coupled to a soft substrate using traction force microscopy. We determine the force exerted on the magnetic bead by the magnet probe - and thus exerted by the magnetic bead on the soft polyacrylamide substrate - as a function of the distance between the probe tip and the magnetic bead. We also show that we can determine the force exerted on a magnetic bead coupled to a cell by the changes in the traction force exerted by the cell on the soft substrate beneath. We thus demonstrate that forces of nanonewton magnitude can be locally exerted on soft substrates or cells and simultaneously determined using traction force microscopy. Application of this method for the in situ measurement of localized exogenous forces exerted on cells can also enable dissection of cellular force transmission pathways.
Collapse
Affiliation(s)
- Joshua Bush
- Mechanical & Aerospace Engineering, Old Dominion University, Kaufman 238e, 1 Old Dominion University, Norfolk, Virginia 23529, USA
| | - Venkat Maruthamuthu
- Mechanical & Aerospace Engineering, Old Dominion University, Kaufman 238e, 1 Old Dominion University, Norfolk, Virginia 23529, USA
| |
Collapse
|
42
|
Multiplexed, high-throughput measurements of cell contraction and endothelial barrier function. J Transl Med 2019; 99:138-145. [PMID: 30310180 PMCID: PMC6309267 DOI: 10.1038/s41374-018-0136-2] [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: 04/17/2018] [Revised: 08/20/2018] [Accepted: 08/31/2018] [Indexed: 12/18/2022] Open
Abstract
Vascular leakage, protein exudation, and edema formation are events commonly triggered by inflammation and facilitated by gaps that form between adjacent endothelial cells (ECs) of the vasculature. In such paracellular gap formation, the role of EC contraction is widely implicated, and even therapeutically targeted. However, related measurement approaches remain slow, tedious, and complex to perform. Here, we have developed a multiplexed, high-throughput screen to simultaneously quantify paracellular gaps, EC contractile forces, and to visualize F-actin stress fibers, and VE-cadherin. As proof-of-principle, we examined barrier-protective mechanisms of the Rho-associated kinase inhibitor, Y-27632, and the canonical agonist of the Tie2 receptor, Angiopoietin-1 (Angpt-1). Y-27632 reduced EC contraction and actin stress fiber formation, whereas Angpt-1 did not. Yet both agents reduced thrombin-, LPS-, and TNFα-induced paracellular gap formation. This unexpected result suggests that Angpt-1 can achieve barrier defense without reducing EC contraction, a mechanism that has not been previously described. This insight was enabled by the multiplex nature of the force-based platform. The high-throughput format we describe should accelerate both mechanistic studies and the screening of pharmacological modulators of endothelial barrier function.
Collapse
|
43
|
Matellan C, Del Río Hernández AE. Where No Hand Has Gone Before: Probing Mechanobiology at the Cellular Level. ACS Biomater Sci Eng 2018; 5:3703-3719. [PMID: 33405886 DOI: 10.1021/acsbiomaterials.8b01206] [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: 01/05/2023]
Abstract
Physical forces and other mechanical stimuli are fundamental regulators of cell behavior and function. Cells are also biomechanically competent: they generate forces to migrate, contract, remodel, and sense their environment. As the knowledge of the mechanisms of mechanobiology increases, the need to resolve and probe increasingly small scales calls for novel technologies to mechanically manipulate cells, examine forces exerted by cells, and characterize cellular biomechanics. Here, we review novel methods to quantify cellular force generation, measure cell mechanical properties, and exert localized piconewton and nanonewton forces on cells, receptors, and proteins. The combination of these technologies will provide further insight on the effect of mechanical stimuli on cells and the mechanisms that convert these stimuli into biochemical and biomechanical activity.
Collapse
Affiliation(s)
- Carlos Matellan
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Armando E Del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
44
|
Bartolák-Suki E, Imsirovic J, Nishibori Y, Krishnan R, Suki B. Regulation of Mitochondrial Structure and Dynamics by the Cytoskeleton and Mechanical Factors. Int J Mol Sci 2017; 18:E1812. [PMID: 28825689 PMCID: PMC5578198 DOI: 10.3390/ijms18081812] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/09/2017] [Accepted: 08/18/2017] [Indexed: 12/21/2022] Open
Abstract
Mitochondria supply cells with energy in the form of ATP, guide apoptosis, and contribute to calcium buffering and reactive oxygen species production. To support these diverse functions, mitochondria form an extensive network with smaller clusters that are able to move along microtubules aided by motor proteins. Mitochondria are also associated with the actin network, which is involved in cellular responses to various mechanical factors. In this review, we discuss mitochondrial structure and function in relation to the cytoskeleton and various mechanical factors influencing cell functions. We first summarize the morphological features of mitochondria with an emphasis on fission and fusion as well as how network properties govern function. We then review the relationship between the mitochondria and the cytoskeletal structures, including mechanical interactions. We also discuss how stretch and its dynamic pattern affect mitochondrial structure and function. Finally, we present preliminary data on how extracellular matrix stiffness influences mitochondrial morphology and ATP generation. We conclude by discussing the more general role that mitochondria may play in mechanobiology and how the mechanosensitivity of mitochondria may contribute to the development of several diseases and aging.
Collapse
Affiliation(s)
| | - Jasmin Imsirovic
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Yuichiro Nishibori
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan.
| | - Ramaswamy Krishnan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| |
Collapse
|