1
|
Dong Y, Wang C, Ding X, Ma X, Huang R, Li M, Yang Q. The characterization of cell traction force on nonflat surfaces with different curvature by elastic hydrogel microspheres. Biotechnol Bioeng 2024; 121:3537-3550. [PMID: 38978386 DOI: 10.1002/bit.28802] [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: 03/20/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
It is of great importance to study the detachment/attachment behaviors of cells (cancer cell, immune cell, and epithelial cell), as they are closely related with tumor metastasis, immunoreaction, and tissue development at variety scales. To characterize the detachment/attachment during the interaction between cells and substrate, some researchers proposed using cell traction force (CTF) as the indicator. To date, various strategies have been developed to measure the CTF. However, these methods only realize the measurements of cell passive forces on flat cases. To quantify the active CTF on nonflat surfaces, which can better mimic the in vivo case, we employed elastic hydrogel microspheres as a force sensor. The microspheres were fabricated by microfluidic chips with controllable size and mechanical properties to mimic substrate. Cells were cultured on microsphere and the CTF led to the deformation of microsphere. By detecting the morphology information, the CTF exerted by attached cells can be calculated by the in-house numerical code. Using these microspheres, the CTF of various cells (including tumor cell, immunological cell, and epithelium cell) were successfully obtained on nonflat surfaces with different curvature radii. The proposed method provides a versatile platform to measure the CTF with high precision and to understand the detachment/attachment behaviors during physiology processes.
Collapse
Affiliation(s)
- Yuqing Dong
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Cong Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xin Ding
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xingquan Ma
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
- School of Civil Engineering and Architecture, Xi'an University of Technology, Xi'an, P.R. China
| | - Rong Huang
- Department of Burn and Plastic Surgery, Second Affiliated Hospital, Air Force Medical University, Xi'an, China
| | - Moxiao Li
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
- Nanjing Center for Multifunctional Lightweight Materials and Structures (MLMS), Nanjing University of Aeronautics and Astronautics, Nanjing, P.R. China
| | - Qingzhen Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
2
|
Schunk CT, Wang W, Sabo LN, Taufalele PV, Reinhart-King CA. Matrix stiffness increases energy efficiency of endothelial cells. Matrix Biol 2024; 133:77-85. [PMID: 39147247 DOI: 10.1016/j.matbio.2024.08.004] [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: 01/11/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/17/2024]
Abstract
To form blood vessels, endothelial cells rearrange their cytoskeleton, generate traction stresses, migrate, and proliferate, all of which require energy. Despite these energetic costs, stiffening of the extracellular matrix promotes tumor angiogenesis and increases cell contractility. However, the interplay between extracellular matrix, cell contractility, and cellular energetics remains mechanistically unclear. Here, we utilized polyacrylamide substrates with various stiffnesses, a real-time biosensor of ATP, and traction force microscopy to show that endothelial cells exhibit increasing traction forces and energy usage trend as substrate stiffness increases. Inhibition of cytoskeleton reorganization via ROCK inhibition resulted in decreased cellular energy efficiency, and an opposite trend was found when cells were treated with manganese to promote integrin affinity. Altogether, our data reveal a link between matrix stiffness, cell contractility, and cell energetics, suggesting that endothelial cells on stiffer substrates can better convert intracellular energy into cellular traction forces. Given the critical role of cellular metabolism in cell function, our study also suggests that not only energy production but also the efficiency of its use plays a vital role in regulating cell behaviors and may help explain how increased matrix stiffness promotes angiogenesis.
Collapse
Affiliation(s)
- Curtis T Schunk
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Lindsey N Sabo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Paul V Taufalele
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | | |
Collapse
|
3
|
Wang H, Jia Z, Fang Y. Chemo-mechanical model of cell polarization initiated by structural polarity. SOFT MATTER 2024; 20:8407-8419. [PMID: 39392308 DOI: 10.1039/d4sm00800f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Cell polarization is crucial in most physiological functions. Living cells at the extracellular matrix (ECM) actively coordinate a polarized morphology to target the preferred signals. In particular, the initial heterogeneity of subcellular components, termed as structural polarity, has been discovered to mediate the early attachment and transmigration of cells in tumour metastasis. However, how heterogeneous cells initiate the early polarization remains incompletely discovered. Here, we establish a multiscale model of a cell to explore the chemo-mechanical mechanisms of cell polarization initiated by structural polarity. The two-dimensional vertex model of the cell is built with the main mechanical components of eukaryotic cells. The initial structural polarity of the modeled cell is introduced by seeding heterogeneous actin filaments at the cell cortex and quantified by the ratio of the filamentous forces at the vertices. Then, the structural polarity is integrated in the reaction-diffusion system of Rho GTPase (Cdc42) at the cell cortex to obtain the traction forces at the leading vertices. Finally, the modeled cell is actuated to spread under the traction forces and discovered to develop into a characteristic polarized morphology. The results indicate that the cell polarization is initiated and dynamically developed by structural polarity through the reaction-diffusion system of Cdc42. In addition, the bistability of Cdc42 activation at the cell cortex is defined and discovered to dominate the polarization status of the cell. Furthermore, biphasic (i.e., positive and negative) durotaxis of the cell is successfully modeled at an ECM with a stiffness gradient, and concluded to be codetermined by the chemo-mechanical coupling of the initial structural polarity and ECM stiffness gradient. The proposed multiscale model provides a quantitative way to probe cell polarization coupled with mechanical stimuli, biochemical reaction and cytoskeletal reorganization, and holds the potential to guide studies of cell polarization under multiple stimuli.
Collapse
Affiliation(s)
- Hexiang Wang
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun, China.
| | - Zhimeng Jia
- College of Automotive Engineering, Jilin University, Changchun, China
| | - Yuqiang Fang
- School of Mechanical and Aerospace Engineering, Jilin University, Changchun, China.
| |
Collapse
|
4
|
Li MJ, Li CX, Li JY, Gong ZC, Shao B, Zhou YC, Xu YJ, Jia MY. Biomechanism of abnormal stress on promoting osteoarthritis of temporomandibular joint through Piezo1 ion channel. J Oral Rehabil 2024; 51:1935-1946. [PMID: 38873703 DOI: 10.1111/joor.13777] [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: 12/18/2023] [Revised: 05/19/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVE This study aimed to investigate whether flow fluid shear stress (FFSS)-mediated signal transduction affects the function of Piezo1 ion channel in chondrocyte and to further explore the role of mechanical overloading in development of temporomandibular joint osteoarthritis (TMJ OA). METHODS Immunohistochemical staining was used to determine the expression of Piezo1 in TMJ OA tissue collected from rat unilateral anterior crossbite (UAC) models. Chondrocytes harvested from normal adult SD rats were treated with FFSS (0, 4, 8, 12 dyn/cm2) in vitro. Immunofluorescent staining, real-time polymerase chain reaction, western blotting, flow cytometry and phalloidin assay were performed to detect the changes of cellular morphology as well as the expression of Piezo1 and certain pro-inflammatory and degradative factors in chondrocyte. RESULTS Immunohistochemical analysis revealed that significantly increased Piezo1 expression was associated with UAC stimulation (p < .05). As applied FFSS escalated (4, 8 and 12 dyn/cm2), the expression levels of Piezo1, ADAMTS-5, MMP-13 and Col-X gradually increased, compared with the non-FFSS group (p < .05). Administering Piezo1 ion channel inhibitor to chondrocytes beforehand, it was observed that expression of ADAMTS-5, MMP-13 and Col-X was substantially decreased following FFSS treatment (p < .05) and the effect of cytoskeletal thinning was counteracted. The activated Piezo1 ion channel enhanced intracellular Ca2+ excess in chondrocytes during abnormal mechanical stimulation and the increased intracellular Ca2+ thinned the cytoskeleton of F-actin. CONCLUSIONS Mechanical overloading activates Piezo1 ion channel to promote pro-inflammation and degradation and to increase Ca2+ concentration in chondrocyte, which may eventually result in TMJ OA.
Collapse
Affiliation(s)
- Meng-Jia Li
- Department of Oral and Maxillofacial Oncology and Surgery, School/Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Chen-Xi Li
- Department of Oral and Maxillofacial Oncology and Surgery, School/Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, School of Stomatology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Yu Li
- Department of Oral and Maxillofacial Oncology and Surgery, School/Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhong-Cheng Gong
- Department of Oral and Maxillofacial Oncology and Surgery, School/Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Bo Shao
- Department of Oral and Maxillofacial Oncology and Surgery, School/Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yu-Chuan Zhou
- Department of Oral and Maxillofacial Oncology and Surgery, School/Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Ying-Jie Xu
- Department of Oral and Maxillofacial Oncology and Surgery, School/Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Meng-Ying Jia
- Department of Oral and Maxillofacial Oncology and Surgery, School/Hospital of Stomatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Stomatological Research Institute of Xinjiang Uygur Autonomous Region, Urumqi, China
| |
Collapse
|
5
|
Blázquez-Carmona P, Ruiz-Mateos R, Barrasa-Fano J, Shapeti A, Martín-Alfonso JE, Domínguez J, Van Oosterwyck H, Reina-Romo E, Sanz-Herrera JA. Quantitative atlas of collagen hydrogels reveals mesenchymal cancer cell traction adaptation to the matrix nanoarchitecture. Acta Biomater 2024; 185:281-295. [PMID: 38992411 DOI: 10.1016/j.actbio.2024.07.002] [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: 02/26/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024]
Abstract
Collagen-based hydrogels are commonly used in mechanobiology to mimic the extracellular matrix. A quantitative analysis of the influence of collagen concentration and properties on the structure and mechanics of the hydrogels is essential for tailored design adjustments for specific in vitro conditions. We combined focused ion beam scanning electron microscopy and rheology to provide a detailed quantitative atlas of the mechanical and nanoscale three-dimensional structural alterations that occur when manipulating different hydrogel's physicochemistry. Moreover, we study the effects of such alterations on the phenotype of breast cancer cells and their mechanical interactions with the extracellular matrix. Regardless of the microenvironment's pore size, porosity or mechanical properties, cancer cells are able to reach a stable mesenchymal-like morphology. Additionally, employing 3D traction force microscopy, a positive correlation between cellular tractions and ECM mechanics is observed up to a critical threshold, beyond which tractions plateau. This suggests that cancer cells in a stable mesenchymal state calibrate their mechanical interactions with the ECM to keep their migration and invasiveness capacities unaltered. STATEMENT OF SIGNIFICANCE: The paper presents a thorough study on the mechanical microenvironment in breast cancer cells during their interaction with collagen based hydrogels of different compositions. The hydrogels' microstructure were obtained using state-of-the-art 3D microscopy, namely focused ion beam-scanning electron microscope (FIB-SEM). FIB-SEM was originally applied in this work to reconstruct complex fibered collagen microstructures within the nanometer range, to obtain key microarchitectural parameters. The mechanical microenvironment of cells was recovered using Traction Force Microscopy (TFM). The obtained results suggest that cells calibrate tractions such that they depend on mechanical, microstructural and physicochemical characteristics of the hydrogels, hence revealing a steric hindrance. We hypothesize that cancer cells studied in this paper tune their mechanical state to keep their migration and invasiveness capacities unaltered.
Collapse
Affiliation(s)
- Pablo Blázquez-Carmona
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - Raquel Ruiz-Mateos
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - Jorge Barrasa-Fano
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Celestijnenlaan 300. B-3001 Heverlee, Belgium
| | - Apeksha Shapeti
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Celestijnenlaan 300. B-3001 Heverlee, Belgium
| | - José Enrique Martín-Alfonso
- Escuela Técnica Superior de Ingeniería, Universidad de Huelva. Avda. de las Fuerzas Armadas s/n, 21007 Huelva, Spain
| | - Jaime Domínguez
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - Hans Van Oosterwyck
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Celestijnenlaan 300. B-3001 Heverlee, Belgium
| | - Esther Reina-Romo
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - José Antonio Sanz-Herrera
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain.
| |
Collapse
|
6
|
Jang I, Menon S, Indra I, Basith R, Beningo KA. Calpain Small Subunit Mediated Secretion of Galectin-3 Regulates Traction Stress. Biomedicines 2024; 12:1247. [PMID: 38927454 PMCID: PMC11200796 DOI: 10.3390/biomedicines12061247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
The complex regulation of traction forces (TF) produced during cellular migration remains poorly understood. We have previously found that calpain 4 (Capn4), the small non-catalytic subunit of the calpain 1 and 2 proteases, regulates the production of TF independent of the proteolytic activity of the larger subunits. Capn4 was later found to facilitate tyrosine phosphorylation and secretion of the lectin-binding protein galectin-3 (Gal3). In this study, recombinant Gal3 (rGal3) was added to the media-enhanced TF generated by capn4-/- mouse embryonic fibroblasts (MEFs). Extracellular Gal3 also rescued defects in the distribution, morphology, and adhesive strength of focal adhesions present in capn4-/- MEF cells. Surprisingly, extracellular Gal3 does not influence mechanosensing. c-Abl kinase was found to affect Gal3 secretion and the production of TF through phosphorylation of Y107 on Gal3. Our study also suggests that Gal3-mediated regulation of TF occurs through signaling pathways triggered by β1 integrin but not by focal adhesion kinase (FAK) Y397 autophosphorylation. Our findings provide insights into the signaling mechanism by which Capn4 and secreted Gal3 regulate cell migration through the modulation of TF distinctly independent from a mechanosensing mechanism.
Collapse
Affiliation(s)
| | | | | | | | - Karen A. Beningo
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA; (I.J.)
| |
Collapse
|
7
|
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
|
8
|
Denisin AK, Kim H, Riedel-Kruse IH, Pruitt BL. Field Guide to Traction Force Microscopy. Cell Mol Bioeng 2024; 17:87-106. [PMID: 38737454 PMCID: PMC11082129 DOI: 10.1007/s12195-024-00801-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/26/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction Traction force microscopy (TFM) is a widely used technique to measure cell contractility on compliant substrates that mimic the stiffness of human tissues. For every step in a TFM workflow, users make choices which impact the quantitative results, yet many times the rationales and consequences for making these decisions are unclear. We have found few papers which show the complete experimental and mathematical steps of TFM, thus obfuscating the full effects of these decisions on the final output. Methods Therefore, we present this "Field Guide" with the goal to explain the mathematical basis of common TFM methods to practitioners in an accessible way. We specifically focus on how errors propagate in TFM workflows given specific experimental design and analytical choices. Results We cover important assumptions and considerations in TFM substrate manufacturing, substrate mechanical properties, imaging techniques, image processing methods, approaches and parameters used in calculating traction stress, and data-reporting strategies. Conclusions By presenting a conceptual review and analysis of TFM-focused research articles published over the last two decades, we provide researchers in the field with a better understanding of their options to make more informed choices when creating TFM workflows depending on the type of cell being studied. With this review, we aim to empower experimentalists to quantify cell contractility with confidence. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00801-6.
Collapse
Affiliation(s)
| | - Honesty Kim
- Department of Bioengineering, Stanford University, Stanford, CA 94305 USA
- Present Address: The Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158 USA
- Department of Molecular and Cellular Biology, and (by courtesy) Departments of Biomedical Engineering, Applied Mathematics, and Physics, University of Arizona, Tucson, AZ 85721 USA
| | - Ingmar H. Riedel-Kruse
- Department of Molecular and Cellular Biology, and (by courtesy) Departments of Biomedical Engineering, Applied Mathematics, and Physics, University of Arizona, Tucson, AZ 85721 USA
| | - Beth L. Pruitt
- Departments of Bioengineering and Mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA 93106 USA
| |
Collapse
|
9
|
Cambria E, Coughlin MF, Floryan MA, Offeddu GS, Shelton SE, Kamm RD. Linking cell mechanical memory and cancer metastasis. Nat Rev Cancer 2024; 24:216-228. [PMID: 38238471 PMCID: PMC11146605 DOI: 10.1038/s41568-023-00656-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 03/01/2024]
Abstract
Metastasis causes most cancer-related deaths; however, the efficacy of anti-metastatic drugs is limited by incomplete understanding of the biological mechanisms that drive metastasis. Focusing on the mechanics of metastasis, we propose that the ability of tumour cells to survive the metastatic process is enhanced by mechanical stresses in the primary tumour microenvironment that select for well-adapted cells. In this Perspective, we suggest that biophysical adaptations favourable for metastasis are retained via mechanical memory, such that the extent of memory is influenced by both the magnitude and duration of the mechanical stress. Among the mechanical cues present in the primary tumour microenvironment, we focus on high matrix stiffness to illustrate how it alters tumour cell proliferation, survival, secretion of molecular factors, force generation, deformability, migration and invasion. We particularly centre our discussion on potential mechanisms of mechanical memory formation and retention via mechanotransduction and persistent epigenetic changes. Indeed, we propose that the biophysical adaptations that are induced by this process are retained throughout the metastatic process to improve tumour cell extravasation, survival and colonization in the distant organ. Deciphering mechanical memory mechanisms will be key to discovering a new class of anti-metastatic drugs.
Collapse
Affiliation(s)
- Elena Cambria
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Mark F Coughlin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marie A Floryan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Giovanni S Offeddu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sarah E Shelton
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
10
|
Emon B, Joy MSH, Lalonde L, Ghrayeb A, Doha U, Ladehoff L, Brockstein R, Saengow C, Ewoldt RH, Saif MTA. Nuclear deformation regulates YAP dynamics in cancer associated fibroblasts. Acta Biomater 2024; 173:93-108. [PMID: 37977292 PMCID: PMC10848212 DOI: 10.1016/j.actbio.2023.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
Cells cultured on stiff 2D substrates exert high intracellular force, resulting in mechanical deformation of their nuclei. This nuclear deformation (ND) plays a crucial role in the transport of Yes Associated Protein (YAP) from the cytoplasm to the nucleus. However, cells in vivo are in soft 3D environment with potentially much lower intracellular forces. Whether and how cells may deform their nuclei in 3D for YAP localization remains unclear. Here, by culturing human colon cancer associated fibroblasts (CAFs) on 2D, 2.5D, and 3D substrates, we differentiated the effects of stiffness, force, and ND on YAP localization. We found that nuclear translocation of YAP depends on the degree of ND irrespective of dimensionality, stiffness and total force. ND induced by the perinuclear force, not the total force, and nuclear membrane curvature correlate strongly with YAP activation. Immunostained slices of human tumors further supported the association between ND and YAP nuclear localization, suggesting ND as a potential biomarker for YAP activation in tumors. Additionally, we conducted quantitative analysis of the force dynamics of CAFs on 2D substrates to construct a stochastic model of YAP kinetics. This model revealed that the probability of YAP nuclear translocation, as well as the residence time in the nucleus follow a power law. This study provides valuable insights into the regulatory mechanisms governing YAP dynamics and highlights the significance of threshold activation in YAP localization. STATEMENT OF SIGNIFICANCE: Yes Associated Protein (YAP), a transcription cofactor, has been identified as one of the drivers of cancer progression. High tumor stiffness is attributed to driving YAP to the nucleus, wherein it activates pro-metastatic genes. Here we show, using cancer associated fibroblasts, that YAP translocation to the nucleus depends on the degree of nuclear deformation, irrespective of stiffness. We also identified that perinuclear force induced membrane curvature correlates strongly with YAP nuclear transport. A novel stochastic model of YAP kinetics unveiled a power law relationship between the activation threshold and persistence time of YAP in the nucleus. Overall, this study provides novel insights into the regulatory mechanisms governing YAP dynamics and the probability of activation that is of immense clinical significance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chaimongkol Saengow
- Mechanical Science & Engineering; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign
| | - Randy H Ewoldt
- Mechanical Science & Engineering; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign
| | - M Taher A Saif
- Mechanical Science & Engineering; Bioengineering; Cancer Center at Illinois.
| |
Collapse
|
11
|
Shu W, Kaplan CN. A multiscale theory for spreading and migration of adhesion-reinforced mesenchymal cells. J R Soc Interface 2023; 20:20230317. [PMID: 38086406 PMCID: PMC10715917 DOI: 10.1098/rsif.2023.0317] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
We present a chemomechanical whole-cell theory for the spreading and migration dynamics of mesenchymal cells that can actively reinforce their adhesion to an underlying viscoelastic substrate as a function of its stiffness. Our multiscale model couples the adhesion reinforcement effect at the subcellular scale with the nonlinear mechanics of the nucleus-cytoskeletal network complex at the cellular scale to explain the concurrent monotonic area-stiffness and non-monotonic speed-stiffness relationships observed in experiments: we consider that large cell spreading on stiff substrates flattens the nucleus, increasing the viscous drag force on it. The resulting force balance dictates a reduction in the migration speed on stiff substrates. We also reproduce the experimental influence of the substrate viscosity on the cell spreading area and migration speed by elucidating how the viscosity may either maintain adhesion reinforcement or prevent it depending on the substrate stiffness. Additionally, our model captures the experimental directed migration behaviour of the adhesion-reinforced cells along a stiffness gradient, known as durotaxis, as well as up or down a viscosity gradient (viscotaxis or anti-viscotaxis), the cell moving towards an optimal viscosity in either case. Overall, our theory explains the intertwined mechanics of the cell spreading, migration speed and direction in the presence of the molecular adhesion reinforcement mechanism.
Collapse
Affiliation(s)
- Wenya Shu
- Department of Physics, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Center for Soft Matter and Biological Physics, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - C. Nadir Kaplan
- Department of Physics, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Center for Soft Matter and Biological Physics, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| |
Collapse
|
12
|
Ngo TKN, Yang SJ, Mao BH, Nguyen TKM, Ng QD, Kuo YL, Tsai JH, Saw SN, Tu TY. A deep learning-based pipeline for analyzing the influences of interfacial mechanochemical microenvironments on spheroid invasion using differential interference contrast microscopic images. Mater Today Bio 2023; 23:100820. [PMID: 37810748 PMCID: PMC10558776 DOI: 10.1016/j.mtbio.2023.100820] [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: 02/18/2023] [Revised: 07/16/2023] [Accepted: 09/24/2023] [Indexed: 10/10/2023] Open
Abstract
Metastasis is the leading cause of cancer-related deaths. During this process, cancer cells are likely to navigate discrete tissue-tissue interfaces, enabling them to infiltrate and spread throughout the body. Three-dimensional (3D) spheroid modeling is receiving more attention due to its strengths in studying the invasive behavior of metastatic cancer cells. While microscopy is a conventional approach for investigating 3D invasion, post-invasion image analysis, which is a time-consuming process, remains a significant challenge for researchers. In this study, we presented an image processing pipeline that utilized a deep learning (DL) solution, with an encoder-decoder architecture, to assess and characterize the invasion dynamics of tumor spheroids. The developed models, equipped with feature extraction and measurement capabilities, could be successfully utilized for the automated segmentation of the invasive protrusions as well as the core region of spheroids situated within interfacial microenvironments with distinct mechanochemical factors. Our findings suggest that a combination of the spheroid culture and DL-based image analysis enable identification of time-lapse migratory patterns for tumor spheroids above matrix-substrate interfaces, thus paving the foundation for delineating the mechanism of local invasion during cancer metastasis.
Collapse
Affiliation(s)
- Thi Kim Ngan Ngo
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Sze Jue Yang
- Department of Artificial Intelligence, Faculty of Computer Science and Information Technology, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Bin-Hsu Mao
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Thi Kim Mai Nguyen
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Qi Ding Ng
- Department of Artificial Intelligence, Faculty of Computer Science and Information Technology, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Yao-Lung Kuo
- Department of Surgery, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
- Department of Surgery, National Cheng Kung University Hospital, Tainan, 70101, Taiwan
| | - Jui-Hung Tsai
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, 70101, Taiwan
| | - Shier Nee Saw
- Department of Artificial Intelligence, Faculty of Computer Science and Information Technology, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ting-Yuan Tu
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, 70101, Taiwan
| |
Collapse
|
13
|
Dazzi C, Mehl J, Benamar M, Gerhardt H, Knaus P, Duda GN, Checa S. External mechanical loading overrules cell-cell mechanical communication in sprouting angiogenesis during early bone regeneration. PLoS Comput Biol 2023; 19:e1011647. [PMID: 37956208 PMCID: PMC10681321 DOI: 10.1371/journal.pcbi.1011647] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 11/27/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Sprouting angiogenesis plays a key role during bone regeneration. For example, insufficient early revascularization of the injured site can lead to delayed or non-healing. During sprouting, endothelial cells are known to be mechano-sensitive and respond to local mechanical stimuli. Endothelial cells interact and communicate mechanically with their surroundings, such as outer-vascular stromal cells, through cell-induced traction forces. In addition, external physiological loads act at the healing site, resulting in tissue deformations and impacting cellular arrangements. How these two distinct mechanical cues (cell-induced and external) impact angiogenesis and sprout patterning in early bone healing remains however largely unknown. Therefore, the aim of this study was to investigate the relative role of externally applied and cell-induced mechanical signals in driving sprout patterning at the onset of bone healing. To investigate cellular self-organisation in early bone healing, an in silico model accounting for the mechano-regulation of sprouting angiogenesis and stromal cell organization was developed. Computer model predictions were compared to in vivo experiments of a mouse osteotomy model stabilized with a rigid or a semirigid fixation system. We found that the magnitude and orientation of principal strains within the healing region can explain experimentally observed sprout patterning, under both fixation conditions. Furthermore, upon simulating the selective inhibition of either cell-induced or externally applied mechanical cues, external mechanical signals appear to overrule the mechanical communication acting on a cell-cell interaction level. Such findings illustrate the relevance of external mechanical signals over the local cell-mediated mechanical cues and could be used in the design of fracture treatment strategies for bone regeneration.
Collapse
Affiliation(s)
- Chiara Dazzi
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Mehl
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Mounir Benamar
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Holger Gerhardt
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Sara Checa
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
14
|
Chen Z, Lv X, Lai L, Xu Y, Zhang F. Effects of riboflavin/ultraviolet-A(UVA) scleral crosslinking on the mechanical behavior of the scleral fibroblasts of lens-induced myopia Guinea pigs. Exp Eye Res 2023; 235:109618. [PMID: 37595677 DOI: 10.1016/j.exer.2023.109618] [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: 12/06/2022] [Revised: 07/25/2023] [Accepted: 08/15/2023] [Indexed: 08/20/2023]
Abstract
Myopia is becoming increasingly severe, and studies have shown that the cellular mechanics of scleral fibroblasts are altered following myopia. Scleral UVA-Riboflavin Collagen Crosslinking(sCXL) is a promising treatment for myopia prevention and control of axial growth. Understanding the mechanical properties of scleral fibroblasts is crucial, as it influences the cellular response and limits the extent of molecular deformation triggered. Thus, our study aimed to investigate the effect of mechanical properties of scleral fibroblasts in a lens-induced myopic guinea pig model following sCXL. For this purpose, we performed the 0.1% riboflavin/UVA scleral crosslinking (365 nm,3 mW/cm2,30 min) in the right eyes of guinea pigs in Group CXL. In Group LIM, the right eyes were only administrated negative lens for 6 weeks. No treatment was performed in both eyes of the guinea pigs in group Control. The scleral fibroblasts were isolated and cultured from the scleral tissue at the cross-linking area in Group CXL and the corresponding area in Group LIM and control. The curve of the length of microtubules inhaled by cells under negative pressure was measured by a microaspiration-based isolation technique, and the equilibrium Young's modulus and apparent viscosity of scleral fibroblasts were calculated by formula fitting. The equilibrium Young's modulus of scleral fibroblasts in group CXL was significantly lower than that in the LIM group (P < 0.01, two-sample t-test between pairs), and there was no significant difference between groups CXL and control. The results show that sCXL can effectively moderate the phenomenon that scleral fibroblasts are not easy to deform after myopia. The apparent viscosity modulus in the CXL group was higher than the groups' control and LIM. Taken together, our data demonstrate the biomechanics of the scleral fibroblasts altered after Riboflavin/UVA scleral collagen cross-linking in a lens-induced myopia model.
Collapse
Affiliation(s)
- Zhe Chen
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Lab, Beijing, China
| | - Xiaotong Lv
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Lab, Beijing, China
| | - Lingbo Lai
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Lab, Beijing, China
| | - Yushan Xu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Lab, Beijing, China
| | - Fengju Zhang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Lab, Beijing, China.
| |
Collapse
|
15
|
SubramanianBalachandar V, Islam MM, Steward RL. A machine learning approach to predict cellular mechanical stresses in response to chemical perturbation. Biophys J 2023; 122:3413-3424. [PMID: 37496269 PMCID: PMC10502424 DOI: 10.1016/j.bpj.2023.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/29/2023] [Accepted: 07/24/2023] [Indexed: 07/28/2023] Open
Abstract
Mechanical stresses generated at the cell-cell level and cell-substrate level have been suggested to be important in a host of physiological and pathological processes. However, the influence various chemical compounds have on the mechanical stresses mentioned above is poorly understood, hindering the discovery of novel therapeutics, and representing a barrier in the field. To overcome this barrier, we implemented two approaches: 1) monolayer boundary predictor and 2) discretized window predictor utilizing either stepwise linear regression or quadratic support vector machine machine learning model to predict the dose-dependent response of tractions and intercellular stresses to chemical perturbation. We used experimental traction and intercellular stress data gathered from samples subject to 0.2 or 2 μg/mL drug concentrations along with cell morphological properties extracted from the bright-field images as predictors to train our model. To demonstrate the predictive capability of our machine learning models, we predicted tractions and intercellular stresses in response to 0 and 1 μg/mL drug concentrations which were not utilized in the training sets. Results revealed the discretized window predictor trained just with four samples (292 images) to best predict both intercellular stresses and tractions using the quadratic support vector machine and stepwise linear regression models, respectively, for the unseen sample images.
Collapse
Affiliation(s)
- VigneshAravind SubramanianBalachandar
- Department of Mechanical and Aerospace Engineering, College of Engineering, University of Central Florida, Orlando, Florida; Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Md Mydul Islam
- Department of Mechanical and Aerospace Engineering, College of Engineering, University of Central Florida, Orlando, Florida
| | - R L Steward
- Department of Mechanical and Aerospace Engineering, College of Engineering, University of Central Florida, Orlando, Florida; Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida.
| |
Collapse
|
16
|
Alonso-Matilla R, Provenzano PP, Odde DJ. Optimal cell traction forces in a generalized motor-clutch model. Biophys J 2023; 122:3369-3385. [PMID: 37475213 PMCID: PMC10465728 DOI: 10.1016/j.bpj.2023.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/25/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023] Open
Abstract
Cells exert forces on mechanically compliant environments to sense stiffness, migrate, and remodel tissue. Cells can sense environmental stiffness via myosin-generated pulling forces acting on F-actin, which is in turn mechanically coupled to the environment via adhesive proteins, akin to a clutch in a drivetrain. In this "motor-clutch" framework, the force transmitted depends on the complex interplay of motor, clutch, and environmental properties. Previous mean-field analysis of the motor-clutch model identified the conditions for optimal stiffness for maximal force transmission via a dimensionless number that combines motor-clutch parameters. However, in this and other previous mean-field analyses, the motor-clutch system is assumed to have balanced motors and clutches and did not consider force-dependent clutch reinforcement and catch bond behavior. Here, we generalize the motor-clutch analytical framework to include imbalanced motor-clutch regimes, with clutch reinforcement and catch bonding, and investigate optimality with respect to all parameters. We found that traction force is strongly influenced by clutch stiffness, and we discovered an optimal clutch stiffness that maximizes traction force, suggesting that cells could tune their clutch mechanical properties to perform a specific function. The results provide guidance for maximizing the accuracy of cell-generated force measurements via molecular tension sensors by designing their mechanosensitive linker peptide to be as stiff as possible. In addition, we found that, on rigid substrates, the mean-field analysis identifies optimal motor properties, suggesting that cells could regulate their myosin repertoire and activity to maximize force transmission. Finally, we found that clutch reinforcement shifts the optimum substrate stiffness to larger values, whereas the optimum substrate stiffness is insensitive to clutch catch bond properties. Overall, our work reveals novel features of the motor-clutch model that can affect the design of molecular tension sensors and provide a generalized analytical framework for predicting and controlling cell adhesion and migration in immunotherapy and cancer.
Collapse
Affiliation(s)
- Roberto Alonso-Matilla
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; University of Minnesota Physical Sciences in Oncology Center, Minneapolis, Minnesota; University of Minnesota Center for Multiparametric Imaging of Tumor Immune Microenvironments, Minneapolis, Minnesota
| | - Paolo P Provenzano
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; University of Minnesota Physical Sciences in Oncology Center, Minneapolis, Minnesota; University of Minnesota Center for Multiparametric Imaging of Tumor Immune Microenvironments, Minneapolis, Minnesota; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota; Department of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota; Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; University of Minnesota Physical Sciences in Oncology Center, Minneapolis, Minnesota; University of Minnesota Center for Multiparametric Imaging of Tumor Immune Microenvironments, Minneapolis, Minnesota; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
17
|
Huang M, Wang H, Mackey C, Chung MC, Guan J, Zheng G, Roy A, Xie M, Vulpe C, Tang X. YAP at the Crossroads of Biomechanics and Drug Resistance in Human Cancer. Int J Mol Sci 2023; 24:12491. [PMID: 37569866 PMCID: PMC10419175 DOI: 10.3390/ijms241512491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 07/30/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Biomechanical forces are of fundamental importance in biology, diseases, and medicine. Mechanobiology is an emerging interdisciplinary field that studies how biological mechanisms are regulated by biomechanical forces and how physical principles can be leveraged to innovate new therapeutic strategies. This article reviews state-of-the-art mechanobiology knowledge about the yes-associated protein (YAP), a key mechanosensitive protein, and its roles in the development of drug resistance in human cancer. Specifically, the article discusses three topics: how YAP is mechanically regulated in living cells; the molecular mechanobiology mechanisms by which YAP, along with other functional pathways, influences drug resistance of cancer cells (particularly lung cancer cells); and finally, how the mechanical regulation of YAP can influence drug resistance and vice versa. By integrating these topics, we present a unified framework that has the potential to bring theoretical insights into the design of novel mechanomedicines and advance next-generation cancer therapies to suppress tumor progression and metastasis.
Collapse
Affiliation(s)
- Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Heyang Wang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Cole Mackey
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32603, USA
| | - Michael C. Chung
- Department of Physics, University of Florida, Gainesville, FL 32611, USA
| | - Juan Guan
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
- Department of Physics, University of Florida, Gainesville, FL 32611, USA
| | - Guangrong Zheng
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL 32603, USA
| | - Arkaprava Roy
- Department of Biostatistics, University of Florida, Gainesville, FL 32603, USA
| | - Mingyi Xie
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32603, USA
| | - Christopher Vulpe
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32603, USA
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
18
|
Ozu M, Galizia L, Alvear-Arias JJ, Fernández M, Caviglia A, Zimmermann R, Guastaferri F, Espinoza-Muñoz N, Sutka M, Sigaut L, Pietrasanta LI, González C, Amodeo G, Garate JA. Mechanosensitive aquaporins. Biophys Rev 2023; 15:497-513. [PMID: 37681084 PMCID: PMC10480384 DOI: 10.1007/s12551-023-01098-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/04/2023] [Indexed: 09/09/2023] Open
Abstract
Cellular systems must deal with mechanical forces to satisfy their physiological functions. In this context, proteins with mechanosensitive properties play a crucial role in sensing and responding to environmental changes. The discovery of aquaporins (AQPs) marked a significant breakthrough in the study of water transport. Their transport capacity and regulation features make them key players in cellular processes. To date, few AQPs have been reported to be mechanosensitive. Like mechanosensitive ion channels, AQPs respond to tension changes in the same range. However, unlike ion channels, the aquaporin's transport rate decreases as tension increases, and the molecular features of the mechanism are unknown. Nevertheless, some clues from mechanosensitive ion channels shed light on the AQP-membrane interaction. The GxxxG motif may play a critical role in the water permeation process associated with structural features in AQPs. Consequently, a possible gating mechanism triggered by membrane tension changes would involve a conformational change in the cytoplasmic extreme of the single file region of the water pathway, where glycine and histidine residues from loop B play a key role. In view of their transport capacity and their involvement in relevant processes related to mechanical forces, mechanosensitive AQPs are a fundamental piece of the puzzle for understanding cellular responses.
Collapse
Affiliation(s)
- Marcelo Ozu
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Biodiversidad y Biología Experimental, Buenos Aires, Argentina
- Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Luciano Galizia
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Biodiversidad y Biología Experimental, Buenos Aires, Argentina
- Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Juan José Alvear-Arias
- Interdisciplinary Center of Neurosciences of Valparaiso, University of Valparaiso, CINV, 2360102 Valparaíso, Chile
- Millennium Nucleus in NanoBioPhysics, Santiago, Chile
| | - Miguel Fernández
- Interdisciplinary Center of Neurosciences of Valparaiso, University of Valparaiso, CINV, 2360102 Valparaíso, Chile
- Millennium Nucleus in NanoBioPhysics, Santiago, Chile
| | - Agustín Caviglia
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Biodiversidad y Biología Experimental, Buenos Aires, Argentina
- Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Rosario Zimmermann
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Biodiversidad y Biología Experimental, Buenos Aires, Argentina
- Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Florencia Guastaferri
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Biodiversidad y Biología Experimental, Buenos Aires, Argentina
- Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Present Address: Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET-UNR), Rosario, Argentina
| | - Nicolás Espinoza-Muñoz
- Interdisciplinary Center of Neurosciences of Valparaiso, University of Valparaiso, CINV, 2360102 Valparaíso, Chile
- Millennium Nucleus in NanoBioPhysics, Santiago, Chile
| | - Moira Sutka
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Biodiversidad y Biología Experimental, Buenos Aires, Argentina
- Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Lorena Sigaut
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Física, Buenos Aires, Argentina
- Instituto de Física de Buenos Aires (IFIBA), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Lía Isabel Pietrasanta
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Física, Buenos Aires, Argentina
- Instituto de Física de Buenos Aires (IFIBA), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Carlos González
- Millennium Nucleus in NanoBioPhysics, Santiago, Chile
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL 33136 USA
- Present Address: Molecular Bioscience Department, University of Texas, Austin, TX 78712 USA
| | - Gabriela Amodeo
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Biodiversidad y Biología Experimental, Buenos Aires, Argentina
- Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - José Antonio Garate
- Interdisciplinary Center of Neurosciences of Valparaiso, University of Valparaiso, CINV, 2360102 Valparaíso, Chile
- Millennium Nucleus in NanoBioPhysics, Santiago, Chile
- Facultad de Ingeniería, Arquitectura y Diseño, Universidad San Sebastián, Bellavista, Santiago, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia y Vida, Universidad San Sebastián, 7750000 Santiago, Chile
| |
Collapse
|
19
|
Rashid SA, Dong Y, Ogasawara H, Vierengel M, Essien ME, Salaita K. All-Covalent Nuclease-Resistant and Hydrogel-Tethered DNA Hairpin Probes Map pN Cell Traction Forces. ACS APPLIED MATERIALS & INTERFACES 2023; 15:33362-33372. [PMID: 37409737 PMCID: PMC10360067 DOI: 10.1021/acsami.3c04826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023]
Abstract
Cells sense and respond to the physical properties of their environment through receptor-mediated signaling, a process known as mechanotransduction, which can modulate critical cellular functions such as proliferation, differentiation, and survival. At the molecular level, cell adhesion receptors, such as integrins, transmit piconewton (pN)-scale forces to the extracellular matrix, and the magnitude of the force plays a critical role in cell signaling. The most sensitive approach to measuring integrin forces involves DNA hairpin-based sensors, which are used to quantify and map forces in living cells. Despite the broad use of DNA hairpin sensors to study a variety of mechanotransduction processes, these sensors are typically anchored to rigid glass slides, which are orders of magnitude stiffer than the extracellular matrix and hence modulate native biological responses. Here, we have developed nuclease-resistant DNA hairpin probes that are all covalently tethered to PEG hydrogels to image cell traction forces on physiologically relevant substrate stiffness. Using HeLa cells as a model cell line, we show that the molecular forces transmitted by integrins are highly sensitive to the bulk modulus of the substrate, and cells cultured on the 6 and 13 kPa gels produced a greater number of hairpin unfolding events compared to the 2 kPa substrates. Tension signals are spatially colocalized with pY118-paxillin, confirming focal adhesion-mediated probe opening. Additionally, we found that integrin forces are greater than 5.8 pN but less than 19 pN on 13 kPa gels. This work provides a general strategy to integrate molecular tension probes into hydrogels, which can better mimic in vivo mechanotransduction.
Collapse
Affiliation(s)
- Sk Aysha Rashid
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Yixiao Dong
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Hiroaki Ogasawara
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Maia Vierengel
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Mark Edoho Essien
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Khalid Salaita
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
20
|
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
|
21
|
Xiao P, Zhang Y, Zeng Y, Yang D, Mo J, Zheng Z, Wang J, Zhang Y, Zhou Z, Zhong X, Yan W. Impaired angiogenesis in ageing: the central role of the extracellular matrix. J Transl Med 2023; 21:457. [PMID: 37434156 PMCID: PMC10334673 DOI: 10.1186/s12967-023-04315-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2023] Open
Abstract
Each step in angiogenesis is regulated by the extracellular matrix (ECM). Accumulating evidence indicates that ageing-related changes in the ECM driven by cellular senescence lead to a reduction in neovascularisation, reduced microvascular density, and an increased risk of tissue ischaemic injury. These changes can lead to health events that have major negative impacts on quality of life and place a significant financial burden on the healthcare system. Elucidating interactions between the ECM and cells during angiogenesis in the context of ageing is neceary to clarify the mechanisms underlying reduced angiogenesis in older adults. In this review, we summarize ageing-related changes in the composition, structure, and function of the ECM and their relevance for angiogenesis. Then, we explore in detail the mechanisms of interaction between the aged ECM and cells during impaired angiogenesis in the older population for the first time, discussing diseases caused by restricted angiogenesis. We also outline several novel pro-angiogenic therapeutic strategies targeting the ECM that can provide new insights into the choice of appropriate treatments for a variety of age-related diseases. Based on the knowledge gathered from recent reports and journal articles, we provide a better understanding of the mechanisms underlying impaired angiogenesis with age and contribute to the development of effective treatments that will enhance quality of life.
Collapse
Affiliation(s)
- Ping Xiao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yuting Zeng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Dehong Yang
- Department of Orthopedics Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiayao Mo
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ziting Zheng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jilei Wang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuxin Zhang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhiyan Zhou
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xincen Zhong
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wenjuan Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
22
|
Subramanian Balachandar VA, Steward RL. Extracellular matrix composition alters endothelial force transmission. Am J Physiol Cell Physiol 2023; 325:C314-C323. [PMID: 37335028 PMCID: PMC10393341 DOI: 10.1152/ajpcell.00106.2023] [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: 03/21/2023] [Revised: 06/02/2023] [Accepted: 06/09/2023] [Indexed: 06/21/2023]
Abstract
Extracellular matrix (ECM) composition is important in a host of pathophysiological processes such as angiogenesis, atherosclerosis, and diabetes, and during each of these processes ECM composition has been reported to change over time. However, the impact ECM composition has on the ability of endothelium to respond mechanically is currently unknown. Therefore, in this study, we seeded human umbilical vein endothelial cells (HUVECs) onto soft hydrogels coated with an ECM concentration of 0.1 mg/mL at the following collagen I (Col-I) and fibronectin (FN) ratios: 100% Col-I, 75% Col-I-25% FN, 50% Col-I-50% FN, 25% Col-I-75% FN, and 100% FN. We subsequently measured tractions, intercellular stresses, strain energy, cell morphology, and cell velocity. Our results revealed that tractions and strain energy are maximal at 50% Col-I-50% FN and minimal at 100% Col-I and 100% FN. Intercellular stress response was maximal on 50% Col-I-50% FN and minimal on 25% Col-I-75% FN. Cell area and cell circularity displayed a divergent relationship for different Col-I and FN ratios. We believe that these results will be of great importance to the cardiovascular field, biomedical field, and cell mechanics.NEW & NOTEWORTHY The endothelium constitutes the innermost layer of all blood vessels and plays an important role in vascular physiology and pathology. During certain vascular diseases, the extracellular matrix has been suggested to transition from a collagen-rich matrix to a fibronectin-rich matrix. In this study, we demonstrate the impact various collagen and fibronectin ratios have on endothelial biomechanical and morphological response.
Collapse
Affiliation(s)
- Vignesh Aravind Subramanian Balachandar
- Department of Mechanical and Aerospace Engineering, College of Engineering and Computer Science, University of Central Florida, Orlando, Florida, United States
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States
| | - Robert L Steward
- Department of Mechanical and Aerospace Engineering, College of Engineering and Computer Science, University of Central Florida, Orlando, Florida, United States
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States
| |
Collapse
|
23
|
Beussman KM, Mollica MY, Leonard A, Miles J, Hocter J, Song Z, Stolla M, Han SJ, Emery A, Thomas WE, Sniadecki NJ. Black dots: High-yield traction force microscopy reveals structural factors contributing to platelet forces. Acta Biomater 2023; 163:302-311. [PMID: 34781024 PMCID: PMC9098698 DOI: 10.1016/j.actbio.2021.11.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/12/2021] [Accepted: 11/10/2021] [Indexed: 01/03/2023]
Abstract
Measuring the traction forces produced by cells provides insight into their behavior and physiological function. Here, we developed a technique (dubbed 'black dots') that microcontact prints a fluorescent micropattern onto a flexible substrate to measure cellular traction forces without constraining cell shape or needing to detach the cells. To demonstrate our technique, we assessed human platelets, which can generate a large range of forces within a population. We find platelets that exert more force have more spread area, are more circular, and have more uniformly distributed F-actin filaments. As a result of the high yield of data obtainable by this technique, we were able to evaluate multivariate mixed effects models with interaction terms and conduct a clustering analysis to identify clusters within our data. These statistical techniques demonstrated a complex relationship between spread area, circularity, F-actin dispersion, and platelet force, including cooperative effects that significantly associate with platelet traction forces. STATEMENT OF SIGNIFICANCE: Cells produce contractile forces during division, migration, or wound healing. Measuring cellular forces provides insight into their health, behavior, and function. We developed a technique that calculates cellular forces by seeding cells onto a pattern and quantifying how much each cell displaces the pattern. This technique is capable of measuring hundreds of cells without needing to detach them. Using this technique to evaluate human platelets, we find that platelets exerting more force tend to have more spread area, are more circular in shape, and have more uniformly distributed cytoskeletal filaments. Due to our high yield of data, we were able to apply statistical techniques that revealed combinatorial effects between these factors.
Collapse
Affiliation(s)
- Kevin M Beussman
- Department of Mechanical Engineering, University of Washington, Seattle, WA, United States
| | - Molly Y Mollica
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Andrea Leonard
- Department of Mechanical Engineering, University of Washington, Seattle, WA, United States
| | - Jeffrey Miles
- Bloodworks Northwest Research Institute, Seattle, WA
| | - John Hocter
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Zizhen Song
- School of Computer Science & Engineering, University of Washington, Seattle, WA, United States
| | - Moritz Stolla
- Bloodworks Northwest Research Institute, Seattle, WA; Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Sangyoon J Han
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, United States
| | - Ashley Emery
- Department of Mechanical Engineering, University of Washington, Seattle, WA, United States
| | - Wendy E Thomas
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Nathan J Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA, United States; Department of Bioengineering, University of Washington, Seattle, WA, United States; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States; Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, United States; Resuscitation Engineering Science Unit (RESCU), University of Washington, Seattle, WA, United States.
| |
Collapse
|
24
|
Ketabat F, Maris T, Duan X, Yazdanpanah Z, Kelly ME, Badea I, Chen X. Optimization of 3D printing and in vitro characterization of alginate/gelatin lattice and angular scaffolds for potential cardiac tissue engineering. Front Bioeng Biotechnol 2023; 11:1161804. [PMID: 37304145 PMCID: PMC10248470 DOI: 10.3389/fbioe.2023.1161804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Background: Engineering cardiac tissue that mimics the hierarchical structure of cardiac tissue remains challenging, raising the need for developing novel methods capable of creating structures with high complexity. Three-dimensional (3D)-printing techniques are among promising methods for engineering complex tissue constructs with high precision. By means of 3D printing, this study aims to develop cardiac constructs with a novel angular structure mimicking cardiac architecture from alginate (Alg) and gelatin (Gel) composite. The 3D-printing conditions were optimized and the structures were characterized in vitro, with human umbilical vein endothelial cells (HUVECs) and cardiomyocytes (H9c2 cells), for potential cardiac tissue engineering. Methods: We synthesized the composites of Alg and Gel with varying concentrations and examined their cytotoxicity with both H9c2 cells and HUVECs, as well as their printability for creating 3D structures of varying fibre orientations (angular design). The 3D-printed structures were characterized in terms of morphology by both scanning electron microscopy (SEM) and synchrotron radiation propagation-based imaging computed tomography (SR-PBI-CT), and elastic modulus, swelling percentage, and mass loss percentage as well. The cell viability studies were conducted via measuring the metabolic activity of the live cells with MTT assay and visualizing the cells with live/dead assay kit. Results: Among the examined composite groups of Alg and Gel, two combinations with ratios of 2 to 1 and 3 to 1 (termed as Alg2Gel1 and Alg3Gel1) showed the highest cell survival; they accordingly were used to fabricate two different structures: a novel angular and a conventional lattice structure. Scaffolds made of Alg3Gel1 showed higher elastic modulus, lower swelling percentage, less mass loss, and higher cell survival compared to that of Alg2Gel1. Although the viability of H9c2 cells and HUVECs on all scaffolds composed of Alg3Gel1 was above 99%, the group of the constructs with the angular design maintained significantly more viable cells compared to other investigated groups. Conclusion: The group of angular 3D-ptinted constructs has illustrated promising properties for cardiac tissue engineering by providing high cell viability for both endothelial and cardiac cells, high mechanical strength as well as appropriate swelling, and degradation properties during 21 days of incubation. Statement of Significance: 3D-printing is an emerging method to create complex constructs with high precision in a large scale. In this study, we have demonstrated that 3D-printing can be used to create compatible constructs from the composite of Alg and Gel with endothelial cells and cardiac cells. Also, we have demonstrated that these constructs are able to enhance the viability of cardiac and endothelial cells via creating a 3D structure mimicking the alignment and orientation of the fibers in the native heart.
Collapse
Affiliation(s)
- Farinaz Ketabat
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Titouan Maris
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Institut Catholique des arts et métiers (ICAM)- Site de Toulouse, Toulouse, France
| | - Xiaoman Duan
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Zahra Yazdanpanah
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Michael E. Kelly
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ildiko Badea
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
25
|
Jetta D, Shireen T, Hua SZ. Epithelial cells sense local stiffness via Piezo1 mediated cytoskeletal reorganization. Front Cell Dev Biol 2023; 11:1198109. [PMID: 37293127 PMCID: PMC10244755 DOI: 10.3389/fcell.2023.1198109] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/10/2023] [Indexed: 06/10/2023] Open
Abstract
Local substrate stiffness is one of the major mechanical inputs for tissue organization during its development and remodeling. It is widely recognized that adherent cells use transmembrane proteins (integrins) at focal adhesions to translate ECM mechanical cues into intracellular bioprocess. Here we show that epithelial cells respond to substrate stiffening primarily via actin cytoskeleton organization, that requires activation of mechanosensitive Piezo1 channels. Piezo1 Knockdown cells eliminated the actin stress fibers that formed on stiff substrates, while it had minimal effect on cell morphology and spreading area. Inhibition of Piezo1 channels with GsMTx4 also significantly reduced stiffness-induced F-actin reorganization, suggesting Piezo1 mediated cation current plays a role. Activation of Piezo1 channels with specific agonist (Yoda1) resulted in thickening of F-actin fibers and enlargement of FAs on stiffer substrates, whereas it did not affect the formation of nascent FAs that facilitate spreading on the soft substrates. These results demonstrate that Piezo1 functions as a force sensor that couples with actin cytoskeleton to distinguish the substrate stiffness and facilitate epithelial adaptive remodeling.
Collapse
Affiliation(s)
- Deekshitha Jetta
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, NY, United States
| | - Tasnim Shireen
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, NY, United States
| | - Susan Z. Hua
- Department of Mechanical and Aerospace Engineering, University at Buffalo, Buffalo, NY, United States
- Department of Physiology and Biophysics, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
26
|
Almeida JA, Mathur J, Lee YL, Sarker B, Pathak A. Mechanically primed cells transfer memory to fibrous matrices for invasion across environments of distinct stiffness and dimensionality. Mol Biol Cell 2023; 34:ar54. [PMID: 36696158 PMCID: PMC10208097 DOI: 10.1091/mbc.e22-10-0469] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/04/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Cells sense and migrate across mechanically dissimilar environments throughout development and disease progression. However, it remains unclear whether mechanical memory of past environments empowers cells to navigate new, three-dimensional extracellular matrices. Here, we show that cells previously primed on stiff, compared with soft, matrices generate a higher level of forces to remodel collagen fibers and promote invasion. This priming advantage persists in dense or stiffened collagen. We explain this memory-dependent, cross-environment cell invasion through a lattice-based model wherein stiff-primed cellular forces remodel collagen and minimize energy required for future cell invasion. According to our model, cells transfer their mechanical memory to the matrix via collagen alignment and tension, and this remodeled matrix informs future cell invasion. Thus, memory-laden cells overcome mechanosensing of softer or challenging future environments via a cell-matrix transfer of memory. Consistent with model predictions, depletion of yes-associated protein destabilizes the cellular memory required for collagen remodeling before invasion. We release tension in collagen fibers via laser ablation and disable fiber remodeling by lysyl-oxidase inhibition, both of which disrupt cell-to-matrix transfer of memory and hamper cross-environment invasion. These results have implications for cancer, fibrosis, and aging, where a potential cell-to-matrix transfer of mechanical memory of cells may generate a prolonged cellular response.
Collapse
Affiliation(s)
- José A. Almeida
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130
| | - Jairaj Mathur
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, MO 63130
| | - Ye Lim Lee
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130
| | - Bapi Sarker
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, MO 63130
| | - Amit Pathak
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, MO 63130
| |
Collapse
|
27
|
Lorentzen KA, Hernanz R, Pinilla E, Nyengaard JR, Wogensen L, Simonsen U. Sex-Dependent Impairment of Endothelium-Dependent Relaxation in Aorta of Mice with Overexpression of Hyaluronan in Tunica Media. Int J Mol Sci 2023; 24:8436. [PMID: 37176139 PMCID: PMC10179165 DOI: 10.3390/ijms24098436] [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: 02/27/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Diabetic macroangiopathy is characterized by increased extracellular matrix deposition, including excessive hyaluronan accumulation, vessel thickening and stiffness, and endothelial dysfunction in large arteries. We hypothesized that the overexpression of hyaluronan in the tunica media also led to endothelial cell (EC) dysfunction. To address this hypothesis, we investigated the following in the aortas of mice with excessive hyaluronan accumulation in the tunica media (HAS-2) and wild-type mice: EC dysfunction via myograph studies, nitric oxide (NO) bioavailability via diaminofluorescence, superoxide formation via dihydroethidium fluorescence, and the distances between ECs via stereological methods. EC dysfunction, characterized by blunted relaxations in response to acetylcholine and decreased NO bioavailability, was found in the aortas of male HAS-2 mice, while it was unaltered in the aortas of female HAS-2 mice. Superoxide levels increased and extracellular superoxide dismutase (ecSOD) expression decreased in the aortas of male and female HAS-2 mice. The EC-EC distances and LDL receptor expression were markedly increased in the HAS-2 aortas of male mice. Our findings suggest hyaluronan increases oxidative stress in the vascular wall and that together with increased EC distance, it is associated with a sex-specific decrease in NO levels and endothelial dysfunction in the aorta of male HAS-2 transgenic mice.
Collapse
Affiliation(s)
- Karen Axelgaard Lorentzen
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University Hospital, Aarhus University, 8000 Aarhus, Denmark
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, 8000 Aarhus, Denmark
| | - Raquel Hernanz
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, 8000 Aarhus, Denmark
- Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28933 Alcorcón, Spain
| | - Estéfano Pinilla
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, 8000 Aarhus, Denmark
| | - Jens Randel Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Center for Stochastic Geometry and Advanced Bioimaging, The Department of Clinical Medicine—Stereology, Aarhus University, 8000 Aarhus, Denmark
| | - Lise Wogensen
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University Hospital, Aarhus University, 8000 Aarhus, Denmark
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
28
|
Ebrahim NA, Mwizerwa ON, Ekwueme EC, Muss TE, Ersland EE, Oba T, Oku K, Nishino M, Hikimoto D, Miyoshi H, Tomotoshi K, Neville CM, Sundback CA. Porous honeycomb film membranes enhance endothelial barrier integrity in human vascular wall bilayer model compared to standard track-etched membranes. J Biomed Mater Res A 2023; 111:701-713. [PMID: 36807502 DOI: 10.1002/jbm.a.37517] [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: 10/06/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/23/2023]
Abstract
In vitro vascular wall bilayer models for drug testing and disease modeling must emulate the physical and biological properties of healthy vascular tissue and its endothelial barrier function. Both endothelial cell (EC)-vascular smooth muscle cell (SMC) interaction across the internal elastic lamina (IEL) and blood vessel stiffness impact endothelial barrier integrity. Polymeric porous track-etched membranes (TEM) typically represent the IEL in laboratory vascular bilayer models. However, TEM stiffness exceeds that of diseased blood vessels, and the membrane pore architecture limits EC-SMC interaction. The mechanical properties of compliant honeycomb film (HCF) membranes better simulate the Young's modulus of healthy blood vessels, and HCFs are thinner (4 vs. 10 μm) and more porous (57 vs. 6.5%) than TEMs. We compared endothelial barrier integrity in vascular wall bilayer models with human ECs and SMCs statically cultured on opposite sides of HCFs and TEMs (5 μm pores) for up to 12 days. Highly segregated localization of tight junction (ZO-1) and adherens junction (VE-cadherin) proteins and quiescent F-actin cytoskeletons demonstrated superior and earlier maturation of interendothelial junctions. Quantifying barrier integrity based on transendothelial electrical resistance (TEER), membranes showed only minor but significant TEER differences despite enhanced junctional protein localization on HCF. Elongated ECs on HCF likely experienced greater paracellular diffusion than blocky ECs on TEM. Also, larger populations of plaques of connexin 43 subunit-containing gap junctions suggested enhanced EC-SMC communication across the more porous, thinner HCF. Compared with standard TEMs, engineered vascular wall bilayers cultured on HCFs better replicate physiologic endothelial barrier integrity.
Collapse
Affiliation(s)
- Neven A Ebrahim
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Olive N Mwizerwa
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Emmanuel C Ekwueme
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Tessa E Muss
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Erik E Ersland
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Takahiro Oba
- Bioscience & Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Keisuke Oku
- Bioscience & Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Masafumi Nishino
- Bioscience & Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Daichi Hikimoto
- Bioscience & Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Hayato Miyoshi
- Bioscience & Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Kimihiko Tomotoshi
- Bioscience & Engineering Laboratories, FUJIFILM Corporation, Kanagawa, Japan
| | - Craig M Neville
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Cathryn A Sundback
- Department of Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
29
|
Cao J, Li H, Tang H, Gu X, Wang Y, Guan D, Du J, Fan Y. Stiff Extracellular Matrix Promotes Invasive Behaviors of Trophoblast Cells. Bioengineering (Basel) 2023; 10:bioengineering10030384. [PMID: 36978775 PMCID: PMC10045595 DOI: 10.3390/bioengineering10030384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/12/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
The effect of extracellular matrix (ECM) stiffness on embryonic trophoblast cells invasion during mammalian embryo implantation remains largely unknown. In this study, we investigated the effects of ECM stiffness on various aspects of human trophoblast cell behaviors during cell-ECM interactions. The mechanical microenvironment of the uterus was simulated by fabricating polyacrylamide (PA) hydrogels with different levels of stiffness. The human choriocarcinoma (JAR) cell lineage was used as the trophoblast model. We found that the spreading area of JAR cells, the formation of focal adhesions, and the polymerization of the F-actin cytoskeleton were all facilitated with increased ECM stiffness. Significantly, JAR cells also exhibited durotactic behavior on ECM with a gradient stiffness. Meanwhile, stiffness of the ECM affects the invasion of multicellular JAR spheroids. These results demonstrated that human trophoblast cells are mechanically sensitive, while the mechanical properties of the uterine microenvironment could play an important role in the implantation process.
Collapse
Affiliation(s)
- Jialing Cao
- Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, Institute of Nanotechnology for Single Cell Analysis, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
- Sino-French Engineer School, Beihang University, Beijing 100083, China
| | - Hangyu Li
- State Key Laboratory of Nonlinear Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongyan Tang
- Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, Institute of Nanotechnology for Single Cell Analysis, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Xuenan Gu
- Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, Institute of Nanotechnology for Single Cell Analysis, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Yan Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Dongshi Guan
- State Key Laboratory of Nonlinear Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Du
- Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, Institute of Nanotechnology for Single Cell Analysis, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, Institute of Nanotechnology for Single Cell Analysis, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| |
Collapse
|
30
|
Mandal K, Sangabathuni S, Haghniaz R, Kawakita S, Mecwan M, Nakayama A, Zhang X, Edalati M, Huang W, Lopez Hernandez A, Jucaud V, Dokmeci MR, Khademhosseini A. Oxygen-generating microparticles downregulate HIF-1α expression, increase cardiac contractility, and mitigate ischemic injury. Acta Biomater 2023; 159:211-225. [PMID: 36669549 PMCID: PMC9992239 DOI: 10.1016/j.actbio.2023.01.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/20/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023]
Abstract
Myocardial hypoxia is the low oxygen tension in the heart tissue implicated in many diseases, including ischemia, cardiac dysfunction, or after heart procurement for transplantation. Oxygen-generating microparticles have recently emerged as a potential strategy for supplying oxygen to sustain cell survival, growth, and tissue functionality in hypoxia. Here, we prepared oxygen-generating microparticles with poly D,L-lactic-co-glycolic acid, and calcium peroxide (CPO), which yielded a continuous morphology capable of sustained oxygen release for up to 24 h. We demonstrated that CPO microparticles increased primary rat cardiomyocyte metabolic activity while not affecting cell viability during hypoxia. Moreover, hypoxia-inducible factor (HIF)-1α, which is upregulated during hypoxia, can be downregulated by delivering oxygen using CPO microparticles. Single-cell traction force microscopy data demonstrated that the reduced energy generated by hypoxic cells could be restored using CPO microparticles. We engineered cardiac tissues that showed higher contractility in the presence of CPO microparticles compared to hypoxic cells. Finally, we observed reduced myocardial injuries in ex vivo rabbit hearts treated with CPO microparticles. In contrast, an acute early myocardial injury was observed for the hearts treated with control saline solution in hypoxia. In conclusion, CPO microparticles improved cell and tissue contractility and gene expression while reducing hypoxia-induced myocardial injuries in the heart. STATEMENT OF SIGNIFICANCE: Oxygen-releasing microparticles can reduce myocardial ischemia, allograft rejection, or irregular heartbeats after heart transplantation. Here we present biodegradable oxygen-releasing microparticles that are capable of sustained oxygen release for more than 24 hrs. We then studied the impact of sustained oxygen release from microparticles on gene expresseion and cardiac cell and tissue function. Previous studies have not measured cardiac tissue or cell mechanics during hypoxia, which is important for understanding proper cardiac function and beating. Using traction force microscopy and an engineered tissue-on-a-chip, we demonstrated that our oxygen-releasing microparticles improve cell and tissue contractility during hypoxia while downregulating the HIF-1α expression level. Finally, using the microparticles, we showed reduced myocardial injuries in rabbit heart tissue, confirming the potential of the particles to be used for organ transplantation or tissue engineering.
Collapse
Affiliation(s)
- Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA.
| | - Sivakoti Sangabathuni
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA; California NanoSystems Institute, University of California, Los Angeles, California 90095, USA
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA; California NanoSystems Institute, University of California, Los Angeles, California 90095, USA; Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Aya Nakayama
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Xuexiang Zhang
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Masoud Edalati
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Wei Huang
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Ana Lopez Hernandez
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Mehmet R Dokmeci
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA.
| |
Collapse
|
31
|
Bontempi M, Capozza R, Visani A, Fini M, Giavaresi G, Gambardella A. Near-Surface Nanomechanics of Medical-Grade PEEK Measured by Atomic Force Microscopy. Polymers (Basel) 2023; 15:polym15030718. [PMID: 36772019 PMCID: PMC9920404 DOI: 10.3390/polym15030718] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/22/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
Detecting subtle changes of surface stiffness at spatial scales and forces relevant to biological processes is crucial for the characterization of biopolymer systems in view of chemical and/or physical surface modification aimed at improving bioactivity and/or mechanical strength. Here, a standard atomic force microscopy setup is operated in nanoindentation mode to quantitatively mapping the near-surface elasticity of semicrystalline polyether ether ketone (PEEK) at room temperature. Remarkably, two localized distributions of moduli at about 0.6 and 0.9 GPa are observed below the plastic threshold of the polymer, at indentation loads in the range of 120-450 nN. This finding is ascribed to the localization of the amorphous and crystalline phases on the free surface of the polymer, detected at an unprecedented level of detail. Our study provides insights to quantitatively characterize complex biopolymer systems on the nanoscale and to guide the optimal design of micro- and nanostructures for advanced biomedical applications.
Collapse
Affiliation(s)
- Marco Bontempi
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Rosario Capozza
- School of Engineering, Institute for Infrastructure and Environment, The University of Edinburgh, Thomas Bayes Road, Edinburgh EH9 3JL, UK
| | - Andrea Visani
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Milena Fini
- Scientific Direction, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Gianluca Giavaresi
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Alessandro Gambardella
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
- Correspondence: ; Tel.: +39-051-636-6513
| |
Collapse
|
32
|
SubramanianBalachandar V, Steward RL. Extracellular Matrix Composition Alters Endothelial Force Transmission. RESEARCH SQUARE 2023:rs.3.rs-2499973. [PMID: 36747754 PMCID: PMC9900979 DOI: 10.21203/rs.3.rs-2499973/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
ECM composition is important in a host of pathophysiological processes such as angiogenesis, atherosclerosis, and diabetes, for example and during each of these processes ECM composition has been reported to change over time. However, the impact ECM composition has on the endothelium’s ability to respond mechanically is currently unknown. Therefore, in this study we seeded human umbilical vein endothelial cells (HUVECs) onto soft hydrogels coated with an ECM concentration of 0.1 mg/mL at the following collagen I (Col-I) and fibronectin (FN) ratios: 100%Col-I, 75%Col-I-25%FN, 50%Col-I-50%FN, 25%Col-I-75%FN, and 100%FN. We subsequently measured tractions, intercellular stresses, strain energy, cell morphology, and cell velocity. Our results revealed huvecs seeded on gels coated with 50% Col-I - 50% FN to have the highest intercellular stresses, tractions, strain energies, but the lowest velocities and cell circularity. Huvecs seeded on 100% Col-I had the lowest tractions, cell area while havingthe highest velocities and cell circularity. In addition, cells cultured on 25% Col-I and 75% FN had the lowest intercellular stresses, but the highest cell area. Huvecs cultured on 100% FN yielded the lowest strain energies. We believe these results will be of great importance to the cardiovascular field, biomedical field, and cell mechanics. Summary: Study the influence of different Col-I - FN ECM compositions on endothelial cell mechanics and morphology.
Collapse
|
33
|
Mohagheghian E, Luo J, Yavitt FM, Wei F, Bhala P, Amar K, Rashid F, Wang Y, Liu X, Ji C, Chen J, Arnold DP, Liu Z, Anseth KS, Wang N. Quantifying stiffness and forces of tumor colonies and embryos using a magnetic microrobot. Sci Robot 2023; 8:eadc9800. [PMID: 36696474 PMCID: PMC10098875 DOI: 10.1126/scirobotics.adc9800] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 12/22/2022] [Indexed: 01/27/2023]
Abstract
Stiffness and forces are two fundamental quantities essential to living cells and tissues. However, it has been a challenge to quantify both 3D traction forces and stiffness (or modulus) using the same probe in vivo. Here, we describe an approach that overcomes this challenge by creating a magnetic microrobot probe with controllable functionality. Biocompatible ferromagnetic cobalt-platinum microcrosses were fabricated, and each microcross (about 30 micrometers) was trapped inside an arginine-glycine-apartic acid-conjugated stiff poly(ethylene glycol) (PEG) round microgel (about 50 micrometers) using a microfluidic device. The stiff magnetic microrobot was seeded inside a cell colony and acted as a stiffness probe by rigidly rotating in response to an oscillatory magnetic field. Then, brief episodes of ultraviolet light exposure were applied to dynamically photodegrade and soften the fluorescent nanoparticle-embedded PEG microgel, whose deformation and 3D traction forces were quantified. Using the microrobot probe, we show that malignant tumor-repopulating cell colonies altered their modulus but not traction forces in response to different 3D substrate elasticities. Stiffness and 3D traction forces were measured, and both normal and shear traction force oscillations were observed in zebrafish embryos from blastula to gastrula. Mouse embryos generated larger tensile and compressive traction force oscillations than shear traction force oscillations during blastocyst. The microrobot probe with controllable functionality via magnetic fields could potentially be useful for studying the mechanoregulation of cells, tissues, and embryos.
Collapse
Affiliation(s)
- Erfan Mohagheghian
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Junyu Luo
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Laboratory for Cellular Biomechanics and Regenerative Medicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - F. Max Yavitt
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
| | - Fuxiang Wei
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Laboratory for Cellular Biomechanics and Regenerative Medicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Parth Bhala
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kshitij Amar
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Fazlur Rashid
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Yuzheng Wang
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Xingchen Liu
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chenyang Ji
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Laboratory for Cellular Biomechanics and Regenerative Medicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Junwei Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Laboratory for Cellular Biomechanics and Regenerative Medicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - David P. Arnold
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Zhen Liu
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
| | - Ning Wang
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
34
|
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
|
35
|
Colasurdo M, Nieves EB, Fernández-Yagüe MA, Franck C, García AJ. Adhesive peptide and polymer density modulate 3D cell traction forces within synthetic hydrogels. Biomaterials 2022; 288:121710. [PMID: 35999082 DOI: 10.1016/j.biomaterials.2022.121710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/27/2022] [Accepted: 07/31/2022] [Indexed: 11/30/2022]
Abstract
Cell-extracellular matrix forces provide pivotal signals regulating diverse physiological and pathological processes. Although mechanobiology has been widely studied in two-dimensional configurations, limited research has been conducted in three-dimensional (3D) systems due to the complex nature of mechanics and cellular behaviors. In this study, we established a platform integrating a well-defined synthetic hydrogel system (PEG-4MAL) with 3D traction force microscopy (TFM) methodologies to evaluate deformation and force responses within synthetic microenvironments, providing insights that are not tractable using biological matrices because of the interdependence of biochemical and biophysical properties and complex mechanics. We dissected the contributions of adhesive peptide density and polymer density, which determines hydrogel stiffness, to 3D force generation for fibroblasts. A critical threshold of adhesive peptide density at a constant matrix elasticity is required for cells to generate 3D forces. Furthermore, matrix displacements and strains decreased with matrix stiffness whereas stresses, and tractions increased with matrix stiffness until reaching constant values at higher stiffness values. Finally, Rho-kinase-dependent contractility and vinculin expression are required to generate significant 3D forces in both collagen and synthetic hydrogels. This research establishes a tunable platform for the study of mechanobiology and provides new insights into how cells sense and transmit forces in 3D.
Collapse
Affiliation(s)
- Mark Colasurdo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Elisa B Nieves
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Marc A Fernández-Yagüe
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Christian Franck
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Andrés J García
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
36
|
Nanoscale geometry determines mechanical biocompatibility of vertically aligned nanofibers. Acta Biomater 2022; 146:235-247. [PMID: 35487425 DOI: 10.1016/j.actbio.2022.04.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 11/24/2022]
Abstract
Vertically aligned carbon nanofibers (VACNFs) are promising material candidates for neural biosensors due to their ability to detect neurotransmitters in physiological concentrations. However, the expected high rigidity of CNFs could induce mechanical mismatch with the brain tissue, eliciting formation of a glial scar around the electrode and thus loss of functionality. We have evaluated mechanical biocompatibility of VACNFs by growing nickel-catalyzed carbon nanofibers of different lengths and inter-fiber distances. Long nanofibers with large inter-fiber distance prevented maturation of focal adhesions, thus constraining cells from obtaining a highly spread morphology that is observed when astrocytes are being contacted with stiff materials commonly used in neural implants. A silicon nanopillar array with 500 nm inter-pillar distance was used to reveal that this inhibition of focal adhesion maturation occurs due to the surface nanoscale geometry, more precisely the inter-fiber distance. Live cell atomic force microscopy was used to confirm astrocytes being significantly softer on the long Ni-CNFs compared to other surfaces, including a soft gelatin hydrogel. We also observed hippocampal neurons to mature and form synaptic contacts when being cultured on both long and short carbon nanofibers, without having to use any adhesive proteins or a glial monoculture, indicating high cytocompatibility of the material also with neuronal population. In contrast, neurons cultured on a planar tetrahedral amorphous carbon sample showed immature neurites and indications of early-stage apoptosis. Our results demonstrate that mechanical biocompatibility of biomaterials is greatly affected by their nanoscale surface geometry, which provides means for controlling how the materials and their mechanical properties are perceived by the cells. STATEMENT OF SIGNIFICANCE: Our research article shows, how nanoscale surface geometry determines mechanical biocompatibility of apparently stiff materials. Specifically, astrocytes were prevented from obtaining highly spread morphology when their adhesion site maturation was inhibited, showing similar morphology on nominally stiff vertically aligned carbon fiber (VACNF) substrates as when being cultured on ultrasoft surfaces. Furthermore, hippocampal neurons matured well and formed synapses on these carbon nanofibers, indicating high biocompatibility of the materials. Interestingly, the same VACNF materials that were used in this study have earlier also been proven to be capable for electrophysiological recordings and sensing neurotransmitters at physiological concentrations with ultra-high sensitivity and selectivity, thus providing a platform for future neural probes or smart culturing surfaces with superior sensing performance and biocompatibility.
Collapse
|
37
|
Aparicio-Yuste R, Muenkel M, Clark AG, Gómez-Benito MJ, Bastounis EE. A Stiff Extracellular Matrix Favors the Mechanical Cell Competition that Leads to Extrusion of Bacterially-Infected Epithelial Cells. Front Cell Dev Biol 2022; 10:912318. [PMID: 35813215 PMCID: PMC9257086 DOI: 10.3389/fcell.2022.912318] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022] Open
Abstract
Cell competition refers to the mechanism whereby less fit cells (“losers”) are sensed and eliminated by more fit neighboring cells (“winners”) and arises during many processes including intracellular bacterial infection. Extracellular matrix (ECM) stiffness can regulate important cellular functions, such as motility, by modulating the physical forces that cells transduce and could thus modulate the output of cellular competitions. Herein, we employ a computational model to investigate the previously overlooked role of ECM stiffness in modulating the forceful extrusion of infected “loser” cells by uninfected “winner” cells. We find that increasing ECM stiffness promotes the collective squeezing and subsequent extrusion of infected cells due to differential cell displacements and cellular force generation. Moreover, we discover that an increase in the ratio of uninfected to infected cell stiffness as well as a smaller infection focus size, independently promote squeezing of infected cells, and this phenomenon is more prominent on stiffer compared to softer matrices. Our experimental findings validate the computational predictions by demonstrating increased collective cell extrusion on stiff matrices and glass as opposed to softer matrices, which is associated with decreased bacterial spread in the basal cell monolayer in vitro. Collectively, our results suggest that ECM stiffness plays a major role in modulating the competition between infected and uninfected cells, with stiffer matrices promoting this battle through differential modulation of cell mechanics between the two cell populations.
Collapse
Affiliation(s)
- Raúl Aparicio-Yuste
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Instituto de Investigación en Ingeniería de Aragón (I3A), University of Zaragoza, Zaragoza, Spain
- Interfaculty Institute of Microbiology and Infection Medicine, Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI, EXC 2124), University of Tübingen, Tübingen, Germany
| | - Marie Muenkel
- Interfaculty Institute of Microbiology and Infection Medicine, Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI, EXC 2124), University of Tübingen, Tübingen, Germany
| | - Andrew G. Clark
- Institute of Cell Biology and Immunology/Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
- Center for Personalized Medicine, University of Tübingen, Tübingen, Germany
| | - María J. Gómez-Benito
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Instituto de Investigación en Ingeniería de Aragón (I3A), University of Zaragoza, Zaragoza, Spain
- *Correspondence: María J. Gómez-Benito, ; Effie E. Bastounis,
| | - Effie E. Bastounis
- Interfaculty Institute of Microbiology and Infection Medicine, Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI, EXC 2124), University of Tübingen, Tübingen, Germany
- *Correspondence: María J. Gómez-Benito, ; Effie E. Bastounis,
| |
Collapse
|
38
|
Pahapale GJ, Tao J, Nikolic M, Gao S, Scarcelli G, Sun SX, Romer LH, Gracias DH. Directing Multicellular Organization by Varying the Aspect Ratio of Soft Hydrogel Microwells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104649. [PMID: 35434926 PMCID: PMC9189654 DOI: 10.1002/advs.202104649] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/08/2022] [Indexed: 06/03/2023]
Abstract
Multicellular organization with precise spatial definition is essential to various biological processes, including morphogenesis, development, and healing in vascular and other tissues. Gradients and patterns of chemoattractants are well-described guides of multicellular organization, but the influences of 3D geometry of soft hydrogels are less well defined. Here, the discovery of a new mode of endothelial cell self-organization guided by combinatorial effects of stiffness and geometry, independent of protein or chemical patterning, is described. Endothelial cells in 2 kPa microwells are found to be ≈30 times more likely to migrate to the edge to organize in ring-like patterns than in stiff 35 kPa microwells. This organization is independent of curvature and significantly more pronounced in 2 kPa microwells with aspect ratio (perimeter/depth) < 25. Physical factors of cells and substrates that drive this behavior are systematically investigated and a mathematical model that explains the organization by balancing the dynamic interaction between tangential cytoskeletal tension, cell-cell, and cell-substrate adhesion is presented. These findings demonstrate the importance of combinatorial effects of geometry and stiffness in complex cellular organization that can be leveraged to facilitate the engineering of bionics and integrated model organoid systems with customized nutrient vascular networks.
Collapse
Affiliation(s)
- Gayatri J. Pahapale
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Jiaxiang Tao
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Milos Nikolic
- Maryland Biophysics ProgramInstitute for Physical Science and TechnologyUniversity of MarylandCollege ParkMD20742USA
| | - Sammy Gao
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Giuliano Scarcelli
- Maryland Biophysics ProgramInstitute for Physical Science and Technology and Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Sean X. Sun
- Department of Mechanical EngineeringCell Biologyand Institute of NanoBioTechnology (INBT)Johns Hopkins UniversityBaltimoreMD21218USA
| | - Lewis H. Romer
- Department of Cell BiologyAnesthesiology and Critical Care MedicineBiomedical EngineeringPediatricsand Center for Cell DynamicsJohns Hopkins School of MedicineBaltimoreMD21205USA
| | - David H. Gracias
- Department of Chemical and Biomolecular EngineeringMaterials Science and EngineeringChemistry and Laboratory for Computational Sensing and Robotics (LCSR)Johns Hopkins UniversityBaltimoreMD21218USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins School of MedicineBaltimoreMD21205USA
| |
Collapse
|
39
|
Yamaguchi N, Knaut H. Focal adhesion-mediated cell anchoring and migration: from in vitro to in vivo. Development 2022; 149:dev200647. [PMID: 35587444 PMCID: PMC9188754 DOI: 10.1242/dev.200647] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cell-extracellular matrix interactions have been studied extensively using cells cultured in vitro. These studies indicate that focal adhesion (FA)-based cell-extracellular matrix interactions are essential for cell anchoring and cell migration. Whether FAs play a similarly important role in vivo is less clear. Here, we summarize the formation and function of FAs in cultured cells and review how FAs transmit and sense force in vitro. Using examples from animal studies, we also describe the role of FAs in cell anchoring during morphogenetic movements and cell migration in vivo. Finally, we conclude by discussing similarities and differences in how FAs function in vitro and in vivo.
Collapse
Affiliation(s)
| | - Holger Knaut
- Skirball Institute of Biomolecular Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
40
|
Rothermel TM, Cook BL, Alford PW. Cellular Microbiaxial Stretching Assay for Measurement and Characterization of the Anisotropic Mechanical Properties of Micropatterned Cells. Curr Protoc 2022; 2:e370. [PMID: 35195953 DOI: 10.1002/cpz1.370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Characterizing the mechanical properties of single cells is important for developing descriptive models of tissue mechanics and improving the understanding of mechanically driven cell processes. Standard methods for measuring single-cell mechanical properties typically provide isotropic mechanical descriptions. However, many cells exhibit specialized geometries in vivo, with anisotropic cytoskeletal architectures reflective of their function, and are exposed to dynamic multiaxial loads, raising the need for more complete descriptions of their anisotropic mechanical properties under complex deformations. Here, we describe the cellular microbiaxial stretching (CμBS) assay in which controlled deformations are applied to micropatterned cells while simultaneously measuring cell stress. CμBS utilizes a set of linear actuators to apply tensile or compressive, short- or long-term deformations to cells micropatterned on a fluorescent bead-doped polyacrylamide gel. Using traction force microscopy principles and the known geometry of the cell and the mechanical properties of the underlying gel, we calculate the stress within the cell to formulate stress-strain curves that can be further used to create mechanical descriptions of the cells, such as strain energy density functions. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Assembly of CμBS stretching constructs Basic Protocol 2: Polymerization of micropatterned, bead-doped polyacrylamide gel on an elastomer membrane Support Protocol: Cell culture and seeding onto CμBS constructs Basic Protocol 3: Implementing CμBS stretching protocols and traction force microscopy Basic Protocol 4: Data analysis and cell stress measurements.
Collapse
Affiliation(s)
- Taylor M Rothermel
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Bernard L Cook
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Patrick W Alford
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
41
|
Swoger M, Gupta S, Charrier EE, Bates M, Hehnly H, Patteson AE. Vimentin Intermediate Filaments Mediate Cell Morphology on Viscoelastic Substrates. ACS APPLIED BIO MATERIALS 2022; 5:552-561. [PMID: 34995457 PMCID: PMC8864613 DOI: 10.1021/acsabm.1c01046] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/27/2021] [Indexed: 12/12/2022]
Abstract
The ability of cells to take and change shape is a fundamental feature underlying development, wound repair, and tissue maintenance. Central to this process is physical and signaling interactions between the three cytoskeletal polymeric networks: F-actin, microtubules, and intermediate filaments (IFs). Vimentin is an IF protein that is essential to the mechanical resilience of cells and regulates cross-talk among the cytoskeleton, but its role in how cells sense and respond to the surrounding extracellular matrix is largely unclear. To investigate vimentin's role in substrate sensing, we designed polyacrylamide hydrogels that mimic the elastic and viscoelastic nature of in vivo tissues. Using wild-type and vimentin-null mouse embryonic fibroblasts, we show that vimentin enhances cell spreading on viscoelastic substrates, even though it has little effect in the limit of purely elastic substrates. Our results provide compelling evidence that vimentin modulates how cells sense and respond to their environment and thus plays a key role in cell mechanosensing.
Collapse
Affiliation(s)
- Maxx Swoger
- Physics
Department, Syracuse University, Syracuse, New York 13244, United States
- BioInspired
Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Sarthak Gupta
- Physics
Department, Syracuse University, Syracuse, New York 13244, United States
- BioInspired
Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Elisabeth E. Charrier
- Institute
of Medicine and Engineering, University
of Pennsylvania, Philadelphia, Pennsylvania 13210, United States
| | - Michael Bates
- Biology
Department, Syracuse University, Syracuse, New York 13244, United States
| | - Heidi Hehnly
- Biology
Department, Syracuse University, Syracuse, New York 13244, United States
| | - Alison E. Patteson
- Physics
Department, Syracuse University, Syracuse, New York 13244, United States
- BioInspired
Institute, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
42
|
Lin C, Zheng X, Lin S, Zhang Y, Wu J, Li Y. Mechanotransduction Regulates the Interplays Between Alveolar Epithelial and Vascular Endothelial Cells in Lung. Front Physiol 2022; 13:818394. [PMID: 35250619 PMCID: PMC8895143 DOI: 10.3389/fphys.2022.818394] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/28/2022] [Indexed: 12/22/2022] Open
Abstract
Mechanical stress plays a critical role among development, functional maturation, and pathogenesis of pulmonary tissues, especially for the alveolar epithelial cells and vascular endothelial cells located in the microenvironment established with vascular network and bronchial-alveolar network. Alveolar epithelial cells are mainly loaded by cyclic strain and air pressure tension. While vascular endothelial cells are exposed to shear stress and cyclic strain. Currently, the emerging evidences demonstrated that non-physiological mechanical forces would lead to several pulmonary diseases, including pulmonary hypertension, fibrosis, and ventilation induced lung injury. Furthermore, a series of intracellular signaling had been identified to be involved in mechanotransduction and participated in regulating the physiological homeostasis and pathophysiological process. Besides, the communications between alveolar epithelium and vascular endothelium under non-physiological stress contribute to the remodeling of the pulmonary micro-environment in collaboration, including hypoxia induced injuries, endothelial permeability impairment, extracellular matrix stiffness elevation, metabolic alternation, and inflammation activation. In this review, we aim to summarize the current understandings of mechanotransduction on the relation between mechanical forces acting on the lung and biological response in mechanical overloading related diseases. We also would like to emphasize the interplays between alveolar epithelium and vascular endothelium, providing new insights into pulmonary diseases pathogenesis, and potential targets for therapy.
Collapse
Affiliation(s)
- Chuyang Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiaolan Zheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Sha Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yue Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jinlin Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Mgharbel A, Migdal C, Bouchonville N, Dupenloup P, Fuard D, Lopez-Soler E, Tomba C, Courçon M, Gulino-Debrac D, Delanoë-Ayari H, Nicolas A. Cells on Hydrogels with Micron-Scaled Stiffness Patterns Demonstrate Local Stiffness Sensing. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:648. [PMID: 35214978 PMCID: PMC8880377 DOI: 10.3390/nano12040648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/31/2022] [Accepted: 02/07/2022] [Indexed: 12/15/2022]
Abstract
Cell rigidity sensing-a basic cellular process allowing cells to adapt to mechanical cues-involves cell capabilities exerting force on the extracellular environment. In vivo, cells are exposed to multi-scaled heterogeneities in the mechanical properties of the surroundings. Here, we investigate whether cells are able to sense micron-scaled stiffness textures by measuring the forces they transmit to the extracellular matrix. To this end, we propose an efficient photochemistry of polyacrylamide hydrogels to design micron-scale stiffness patterns with kPa/µm gradients. Additionally, we propose an original protocol for the surface coating of adhesion proteins, which allows tuning the surface density from fully coupled to fully independent of the stiffness pattern. This evidences that cells pull on their surroundings by adjusting the level of stress to the micron-scaled stiffness. This conclusion was achieved through improvements in the traction force microscopy technique, e.g., adapting to substrates with a non-uniform stiffness and achieving a submicron resolution thanks to the implementation of a pyramidal optical flow algorithm. These developments provide tools for enhancing the current understanding of the contribution of stiffness alterations in many pathologies, including cancer.
Collapse
Affiliation(s)
- Abbas Mgharbel
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
- University Grenoble Alps, CEA, CNRS, Inserm, BIG-BCI, 38000 Grenoble, France; (M.C.); (D.G.-D.)
| | - Camille Migdal
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
- University Grenoble Alps, CEA, CNRS, Inserm, BIG-BCI, 38000 Grenoble, France; (M.C.); (D.G.-D.)
| | - Nicolas Bouchonville
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
| | - Paul Dupenloup
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
| | - David Fuard
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
| | - Eline Lopez-Soler
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
- University Grenoble Alps, CEA, CNRS, Inserm, BIG-BCI, 38000 Grenoble, France; (M.C.); (D.G.-D.)
| | - Caterina Tomba
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
- University Grenoble Alps, CNRS, Grenoble INP, Institut Néel, 38000 Grenoble, France
| | - Marie Courçon
- University Grenoble Alps, CEA, CNRS, Inserm, BIG-BCI, 38000 Grenoble, France; (M.C.); (D.G.-D.)
| | - Danielle Gulino-Debrac
- University Grenoble Alps, CEA, CNRS, Inserm, BIG-BCI, 38000 Grenoble, France; (M.C.); (D.G.-D.)
| | - Héléne Delanoë-Ayari
- Université de Lyon, University Claude Bernard Lyon1, CNRS, Institut Lumière Matière, 69622 Villeurbanne, France;
| | - Alice Nicolas
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
| |
Collapse
|
44
|
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
|
45
|
Tamura A, Lee DH, Arisaka Y, Kang TW, Yui N. Post-Cross-Linking of Collagen Hydrogels by Carboxymethylated Polyrotaxanes for Simultaneously Improving Mechanical Strength and Cell Proliferation. ACS Biomater Sci Eng 2022; 8:588-597. [PMID: 34994537 DOI: 10.1021/acsbiomaterials.1c01521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
To improve the mechanical properties of collagen hydrogels, which are widely utilized as biomaterials, post-cross-linking of collagen hydrogels was performed using polyrotaxane (PRX) as a cross-linker. Herein, carboxymethyl group-modified PRXs (CMPRs) composed of carboxymethylated α-cyclodextrins (α-CDs) threaded along poly(ethylene glycol) (PEG) capped with bulky stoppers were used to cross-link via reaction with the amino groups in the collagen. Four series of CMPRs with different α-CD threading ratios and axle PEG molecular weights were used for the post-cross-linking of the collagen hydrogels to verify the optimal CMPR chemical compositions. The post-cross-linking of the collagen hydrogels with CMPRs improved the swelling ratios and mechanical properties, such as viscoelasticity and tensile strength. Among the tested CMPRs, CMPRs with an axle PEG molecular weight of 35,000 (PEG35k) resulted in better mechanical properties than CMPRs with a PEG10k axis. Additionally, the cell adhesion and proliferation were greatly improved on the surface of the collagen hydrogels post-cross-linked with CMPRs with the PEG35k axle. These findings suggest that the molecular weight of an axle polymer in CMPRs is a more important parameter than the α-CD threading ratios. Accordingly, the post-cross-linking of hydrogels with PRXs is promising for improving the mechanical properties and biomaterial functions of collagen hydrogels.
Collapse
Affiliation(s)
- Atsushi Tamura
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| | - Dae Hoon Lee
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan.,Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Yoshinori Arisaka
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| | - Tae Woong Kang
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| | - Nobuhiko Yui
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| |
Collapse
|
46
|
Panzetta V, Musella I, Fusco S, Netti PA. ECM Mechanoregulation in Malignant Pleural Mesothelioma. Front Bioeng Biotechnol 2022; 10:797900. [PMID: 35237573 PMCID: PMC8883334 DOI: 10.3389/fbioe.2022.797900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/05/2022] [Indexed: 01/16/2023] Open
Abstract
Malignant pleural mesothelioma is a relatively rare, but devastating tumor, because of the difficulties in providing early diagnosis and effective treatments with conventional chemo- and radiotherapies. Patients usually present pleural effusions that can be used for diagnostic purposes by cytological analysis. This effusion cytology may take weeks or months to establish and has a limited sensitivity (30%-60%). Then, it is becoming increasingly urgent to develop alternative investigative methods to support the diagnosis of mesothelioma at an early stage when this cancer can be treated successfully. To this purpose, mechanobiology provides novel perspectives into the study of tumor onset and progression and new diagnostic tools for the mechanical characterization of tumor tissues. Here, we report a mechanical and biophysical characterization of malignant pleural mesothelioma cells as additional support to the diagnosis of pleural effusions. In particular, we examined a normal mesothelial cell line (Met5A) and two epithelioid mesothelioma cell lines (REN and MPP89), investigating how malignant transformation can influence cellular function like proliferation, cell migration, and cell spreading area with respect to the normal ones. These alterations also correlated with variations in cytoskeletal mechanical properties that, in turn, were measured on substrates mimicking the stiffness of patho-physiological ECM.
Collapse
Affiliation(s)
- Valeria Panzetta
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
- Centro di Ricerca Interdipartimentale sui Biomateriali CRIB, University of Naples Federico II, Naples, Italy
- Istituto Italiano di Tecnologia, IIT@CRIB, Naples, Italy
| | - Ida Musella
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
- Istituto Italiano di Tecnologia, IIT@CRIB, Naples, Italy
| | - Sabato Fusco
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Paolo A. Netti
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Naples, Italy
- Centro di Ricerca Interdipartimentale sui Biomateriali CRIB, University of Naples Federico II, Naples, Italy
- Istituto Italiano di Tecnologia, IIT@CRIB, Naples, Italy
| |
Collapse
|
47
|
Liang C, Huang M, Li T, Li L, Sussman H, Dai Y, Siemann DW, Xie M, Tang X. Towards an integrative understanding of cancer mechanobiology: calcium, YAP, and microRNA under biophysical forces. SOFT MATTER 2022; 18:1112-1148. [PMID: 35089300 DOI: 10.1039/d1sm01618k] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
An increasing number of studies have demonstrated the significant roles of the interplay between microenvironmental mechanics in tissues and biochemical-genetic activities in resident tumor cells at different stages of tumor progression. Mediated by molecular mechano-sensors or -transducers, biomechanical cues in tissue microenvironments are transmitted into the tumor cells and regulate biochemical responses and gene expression through mechanotransduction processes. However, the molecular interplay between the mechanotransduction processes and intracellular biochemical signaling pathways remains elusive. This paper reviews the recent advances in understanding the crosstalk between biomechanical cues and three critical biochemical effectors during tumor progression: calcium ions (Ca2+), yes-associated protein (YAP), and microRNAs (miRNAs). We address the molecular mechanisms underpinning the interplay between the mechanotransduction pathways and each of the three effectors. Furthermore, we discuss the functional interactions among the three effectors in the context of soft matter and mechanobiology. We conclude by proposing future directions on studying the tumor mechanobiology that can employ Ca2+, YAP, and miRNAs as novel strategies for cancer mechanotheraputics. This framework has the potential to bring insights into the development of novel next-generation cancer therapies to suppress and treat tumors.
Collapse
Affiliation(s)
- Chenyu Liang
- Department of Mechanical & Aerospace Engineering, Herbert Wertheim College of Engineering (HWCOE), Gainesville, FL, 32611, USA.
- UF Health Cancer Center (UFHCC), Gainesville, FL, 32611, USA
| | - Miao Huang
- Department of Mechanical & Aerospace Engineering, Herbert Wertheim College of Engineering (HWCOE), Gainesville, FL, 32611, USA.
- UF Health Cancer Center (UFHCC), Gainesville, FL, 32611, USA
| | - Tianqi Li
- UF Health Cancer Center (UFHCC), Gainesville, FL, 32611, USA
- Department of Biochemistry and Molecular Biology, College of Medicine (COM), Gainesville, FL, 32611, USA.
| | - Lu Li
- UF Health Cancer Center (UFHCC), Gainesville, FL, 32611, USA
- Department of Biochemistry and Molecular Biology, College of Medicine (COM), Gainesville, FL, 32611, USA.
| | - Hayley Sussman
- Department of Radiation Oncology, COM, Gainesville, FL, 32611, USA
| | - Yao Dai
- UF Health Cancer Center (UFHCC), Gainesville, FL, 32611, USA
- UF Genetics Institute (UFGI), University of Florida (UF), Gainesville, FL, 32611, USA
| | - Dietmar W Siemann
- UF Health Cancer Center (UFHCC), Gainesville, FL, 32611, USA
- UF Genetics Institute (UFGI), University of Florida (UF), Gainesville, FL, 32611, USA
| | - Mingyi Xie
- UF Health Cancer Center (UFHCC), Gainesville, FL, 32611, USA
- Department of Biochemistry and Molecular Biology, College of Medicine (COM), Gainesville, FL, 32611, USA.
- Department of Biomedical Engineering, College of Engineering (COE), University of Delaware (UD), Newark, DE, 19716, USA
| | - Xin Tang
- Department of Mechanical & Aerospace Engineering, Herbert Wertheim College of Engineering (HWCOE), Gainesville, FL, 32611, USA.
- UF Health Cancer Center (UFHCC), Gainesville, FL, 32611, USA
| |
Collapse
|
48
|
Cheung BCH, Hodgson L, Segall JE, Wu M. Spatial and temporal dynamics of RhoA activities of single breast tumor cells in a 3D environment revealed by a machine learning-assisted FRET technique. Exp Cell Res 2022; 410:112939. [PMID: 34813733 PMCID: PMC8714707 DOI: 10.1016/j.yexcr.2021.112939] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 01/17/2023]
Abstract
One of the hallmarks of cancer cells is their exceptional ability to migrate within the extracellular matrix (ECM) for gaining access to the circulatory system, a critical step of cancer metastasis. RhoA, a small GTPase, is known to be a key molecular switch that toggles between actomyosin contractility and lamellipodial protrusion during cell migration. Current understanding of RhoA activity in cell migration has been largely derived from studies of cells plated on a two-dimensional (2D) substrate using a FRET biosensor. There has been increasing evidence that cells behave differently in a more physiologically relevant three-dimensional (3D) environment. However, studies of RhoA activities in 3D have been hindered by low signal-to-noise ratio in fluorescence imaging. In this paper, we present a a machine learning-assisted FRET technique to follow the spatiotemporal dynamics of RhoA activities of single breast tumor cells (MDA-MB-231) migrating in a 3D as well as a 2D environment. We found that RhoA activity is more polarized along the long axis of the cell for single cells migrating on 2D fibronectin-coated glass versus those embedded in 3D collagen matrices. In particular, RhoA activities of cells in 2D exhibit a distinct front-to-back and back-to-front movement during migration in contrast to those in 3D. Finally, regardless of dimensionality, RhoA polarization is found to be moderately correlated with cell shape.
Collapse
Affiliation(s)
- Brian CH Cheung
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Louis Hodgson
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA,Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jeffrey E Segall
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mingming Wu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA,Corresponding author:
| |
Collapse
|
49
|
Cecchini A, Cornelison DDW. Eph/Ephrin-Based Protein Complexes: The Importance of cis Interactions in Guiding Cellular Processes. Front Mol Biosci 2022; 8:809364. [PMID: 35096972 PMCID: PMC8793696 DOI: 10.3389/fmolb.2021.809364] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Although intracellular signal transduction is generally represented as a linear process that transmits stimuli from the exterior of a cell to the interior via a transmembrane receptor, interactions with additional membrane-associated proteins are often critical to its success. These molecules play a pivotal role in mediating signaling via the formation of complexes in cis (within the same membrane) with primary effectors, particularly in the context of tumorigenesis. Such secondary effectors may act to promote successful signaling by mediating receptor-ligand binding, recruitment of molecular partners for the formation of multiprotein complexes, or differential signaling outcomes. One signaling family whose contact-mediated activity is frequently modulated by lateral interactions at the cell surface is Eph/ephrin (EphA and EphB receptor tyrosine kinases and their ligands ephrin-As and ephrin-Bs). Through heterotypic interactions in cis, these molecules can promote a diverse range of cellular activities, including some that are mutually exclusive (cell proliferation and cell differentiation, or adhesion and migration). Due to their broad expression in most tissues and their promiscuous binding within and across classes, the cellular response to Eph:ephrin interaction is highly variable between cell types and is dependent on the cellular context in which binding occurs. In this review, we will discuss interactions between molecules in cis at the cell membrane, with emphasis on their role in modulating Eph/ephrin signaling.
Collapse
Affiliation(s)
- Alessandra Cecchini
- Division of Biological Sciences, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - D. D. W. Cornelison
- Division of Biological Sciences, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
- *Correspondence: D. D. W. Cornelison,
| |
Collapse
|
50
|
Kreysing E, Hugh JM, Foster SK, Andresen K, Greenhalgh RD, Pillai EK, Dimitracopoulos A, Keyser UF, Franze K. Effective cell membrane tension is independent of polyacrylamide substrate stiffness. PNAS NEXUS 2022; 2:pgac299. [PMID: 36733291 PMCID: PMC9887938 DOI: 10.1093/pnasnexus/pgac299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 01/09/2023] [Indexed: 01/11/2023]
Abstract
Most animal cells are surrounded by a cell membrane and an underlying actomyosin cortex. Both structures are linked, and they are under tension. In-plane membrane tension and cortical tension both influence many cellular processes, including cell migration, division, and endocytosis. However, while actomyosin tension is regulated by substrate stiffness, how membrane tension responds to mechanical substrate properties is currently poorly understood. Here, we probed the effective membrane tension of neurons and fibroblasts cultured on glass and polyacrylamide substrates of varying stiffness using optical tweezers. In contrast to actomyosin-based traction forces, both peak forces and steady-state tether forces of cells cultured on hydrogels were independent of substrate stiffness and did not change after blocking myosin II activity using blebbistatin, indicating that tether and traction forces are not directly linked. Peak forces in fibroblasts on hydrogels were about twice as high as those in neurons, indicating stronger membrane-cortex adhesion in fibroblasts. Steady-state tether forces were generally higher in cells cultured on hydrogels than on glass, which we explain by a mechanical model. Our results provide new insights into the complex regulation of effective membrane tension and pave the way for a deeper understanding of the biological processes it instructs.
Collapse
Affiliation(s)
| | | | - Sarah K Foster
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK,Systems Biology of Microbial Communities, Cluster of Excellence—CMFI, University of Tübingen, 72076 Tübingen, Germany
| | - Kurt Andresen
- Department of Physics, Gettysburg College, Gettysburg, PA 17325, USA
| | - Ryan D Greenhalgh
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Eva K Pillai
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Andrea Dimitracopoulos
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | | | | |
Collapse
|