1
|
Wang Y, Strauss S, Smith RS, Sampathkumar A. Actin-mediated avoidance of tricellular junction influences global topology at the Arabidopsis shoot apical meristem. Cell Rep 2024; 43:114844. [PMID: 39418163 DOI: 10.1016/j.celrep.2024.114844] [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: 04/03/2024] [Revised: 07/22/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Division plane orientation contributes to cell shape and topological organization, playing a key role in morphogenesis, but the precise physical and molecular mechanism influencing these processes remains largely obscure in plants. In particular, it is less clear how the placement of the new walls occurs in relation to the walls of neighboring cells. Here, we show that genetic perturbation of the actin cytoskeleton results in more rectangular cell shapes and higher incidences of four-way junctions, perturbing the global topology of cells in the shoot apical meristem of Arabidopsis thaliana. Actin mutants also exhibit changes in the expansion rate of the new versus the maternal cell wall after division, affecting the evolution of internal angles at tricellular junctions. Further, the increased width of the preprophase band in the actin mutant contributes to inaccuracy in the placement of the new cell wall. Computational simulation further substantiates this hypothesis and reproduces the observed cell shape defects.
Collapse
Affiliation(s)
- Yang Wang
- Max Planck Institute of Molecular Plant Physiology, Am Muehlenberg 1, 14476 Potsdam, Germany; Department of Plant and Environmental Science, University of Copenhagen, Thorvaldsensvej 40, 1871 Copenhagen, Denmark
| | - Soeren Strauss
- Department of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, Carl-von-Linné-Weg 10, 50829 Cologne, Germany
| | - Richard S Smith
- Department of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, Carl-von-Linné-Weg 10, 50829 Cologne, Germany; Department of Computational and Systems Biology, John Innes Centre, Norwich Research Park, Colney Ln, NR4 7UH Norwich, UK
| | - Arun Sampathkumar
- Max Planck Institute of Molecular Plant Physiology, Am Muehlenberg 1, 14476 Potsdam, Germany.
| |
Collapse
|
2
|
Delgado D, Desroches S, Kang G, Aldabii Y, Harris AR. An open-source combined atomic force microscope and optical microscope for mechanobiology studies. Heliyon 2024; 10:e38214. [PMID: 39386786 PMCID: PMC11462344 DOI: 10.1016/j.heliyon.2024.e38214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/29/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Atomic Force Microscopy (AFM) has become the gold standard tool for measuring mechanical properties of biological samples including proteins, single cells and tissues. However, investment in this specialized equipment and gaining expertise in its operation are significant obstacles for non-experts looking to adopt this technique. To address this, we have designed an AFM based mechanical measurement system for measuring cell mechanical properties which is combined with a custom inverted fluorescence microscope which can be used for characterizing mechanosensitive responses. This system, through its ease of use and low setup cost, will promote interdisciplinary research leading to new insights into the role of cell mechanics and mechanosensitive responses in physiology and disease.
Collapse
Affiliation(s)
- Daniel Delgado
- Department of Mechanical and Aerospace Engineering, Carleton University, 1125 Colonel by Drive, Ottawa, Ontario, Canada, K1S 5B6
| | - Sarah Desroches
- Department of Mechanical and Aerospace Engineering, Carleton University, 1125 Colonel by Drive, Ottawa, Ontario, Canada, K1S 5B6
- Ottawa-Carleton Institute for Biomedical Engineering Graduate Program, Carleton University, 1125 Colonel by Drive, Ottawa, Ontario, Canada, K1S 5B6
| | - Gia Kang
- Department of Mechanical and Aerospace Engineering, Carleton University, 1125 Colonel by Drive, Ottawa, Ontario, Canada, K1S 5B6
| | - Yousef Aldabii
- Department of Mechanical and Aerospace Engineering, Carleton University, 1125 Colonel by Drive, Ottawa, Ontario, Canada, K1S 5B6
| | - Andrew R. Harris
- Department of Mechanical and Aerospace Engineering, Carleton University, 1125 Colonel by Drive, Ottawa, Ontario, Canada, K1S 5B6
| |
Collapse
|
3
|
Wang J, Li M, Wu W, Zhang H, Yang Y, Usman M, Aernouts B, Loor JJ, Xu C. Inflammatory Signaling via PEIZO1 Engages and Enhances the LPS-Mediated Apoptosis during Clinical Mastitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20321-20330. [PMID: 39229907 DOI: 10.1021/acs.jafc.4c04421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Bovine clinical mastitis is characterized by inflammation and immune responses, with apoptosis of mammary epithelial cells as a cellular reaction to infection. PIEZO1, identified as a mechanotransduction effector channel in nonruminant animals and sensitive to both mechanical stimuli or inflammatory signals like lipopolysaccharide (LPS). However, its role in inflammatory processes in cattle has not been well-documented. The aim of this study was to elucidate the in situ expression of PIEZO1 in bovine mammary gland and its potential involvement in clinical mastitis. We observed widespread distribution and upregulation of PIEZO1 in mammary epithelial cells in clinical mastitis cows and LPS-induced mouse models, indicating a conserved role across species. In vitro studies using mammary epithelial cells (MAC-T) revealed that LPS upregulates PIEZO1. Notably, the effects of PIEZO1 artificial activator Yoda1 increased apoptosis and NLRP3 expression, effects mitigated by PIEZO1 silencing or NLRP3 inhibition. In conclusion, the activation of the PIEZO1-NLRP3 pathway induces abnormal apoptosis in mammary epithelial cells, potentially serving as a regulatory mechanism to combat inflammatory responses to abnormal stimuli.
Collapse
Affiliation(s)
- Jingyi Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Ming Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Wenda Wu
- School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230009, China
| | - HuiJing Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Yue Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| | - Muhammad Usman
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Ben Aernouts
- Department of Biosystems, Division of Animal and Human Health Engineering, Faculty of Engineering Technology, KU Leuven University, Campus Geel, Kleinhoefstraat 4, 2440 Geel, Belgium
| | - Juan J Loor
- Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Chuang Xu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Yuanmingyuan West Road, Beijing 100193, China
| |
Collapse
|
4
|
Mowla A, Hepburn MS, Li J, Vahala D, Amos SE, Hirvonen LM, Sanderson RW, Wijesinghe P, Maher S, Choi YS, Kennedy BF. Multimodal mechano-microscopy reveals mechanical phenotypes of breast cancer spheroids in three dimensions. APL Bioeng 2024; 8:036113. [PMID: 39257700 PMCID: PMC11387014 DOI: 10.1063/5.0213077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/01/2024] [Indexed: 09/12/2024] Open
Abstract
Cancer cell invasion relies on an equilibrium between cell deformability and the biophysical constraints imposed by the extracellular matrix (ECM). However, there is little consensus on the nature of the local biomechanical alterations in cancer cell dissemination in the context of three-dimensional (3D) tumor microenvironments (TMEs). While the shortcomings of two-dimensional (2D) models in replicating in situ cell behavior are well known, 3D TME models remain underutilized because contemporary mechanical quantification tools are limited to surface measurements. Here, we overcome this major challenge by quantifying local mechanics of cancer cell spheroids in 3D TMEs. We achieve this using multimodal mechano-microscopy, integrating optical coherence microscopy-based elasticity imaging with confocal fluorescence microscopy. We observe that non-metastatic cancer spheroids show no invasion while showing increased peripheral cell elasticity in both stiff and soft environments. Metastatic cancer spheroids, however, show ECM-mediated softening in a stiff microenvironment and, in a soft environment, initiate cell invasion with peripheral softening associated with early metastatic dissemination. This exemplar of live-cell 3D mechanotyping supports that invasion increases cell deformability in a 3D context, illustrating the power of multimodal mechano-microscopy for quantitative mechanobiology in situ.
Collapse
Affiliation(s)
| | | | | | - Danielle Vahala
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Sebastian E Amos
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Liisa M Hirvonen
- Centre for Microscopy, Characterisation and Analysis, The University of Western Australia, Perth, WA 6009, Australia
| | | | - Philip Wijesinghe
- Centre of Biophotonics, SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews KY16 9SS, United Kingdom
| | - Samuel Maher
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | | |
Collapse
|
5
|
Niraula G, Pyne A, Wang X. Develop Tandem Tension Sensor to Gauge Integrin-Transmitted Molecular Forces. ACS Sens 2024; 9:3660-3670. [PMID: 38968930 PMCID: PMC11287754 DOI: 10.1021/acssensors.4c00756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/11/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024]
Abstract
DNA-based tension sensors have innovated the imaging and calibration of mechanosensitive receptor-transmitted molecular forces, such as integrin tensions. However, these sensors mainly serve as binary reporters, only indicating if molecular forces exceed one predefined threshold. Here, we have developed tandem tension sensor (TTS), which comprises two consecutive force-sensing units, each with unique force detection thresholds and distinct fluorescence spectra, thereby enabling the quantification of molecular forces with dual reference levels. With TTS, we revealed that vinculin is not required for transmitting integrin tensions at approximately 10 pN (piconewtons) but is essential for elevating integrin tensions beyond 20 pN in focal adhesions (FAs). Such high tensions have emerged during the early stage of FA formation. TTS also successfully detected changes in integrin tensions in response to disrupted actin formation, inhibited myosin activity, and tuned substrate elasticity. We also applied TTS to examine integrin tensions in platelets and revealed two force regimes, with integrin tensions surpassing 20 pN at cell central regions and 13-20 pN integrin tensions at the cell edge. Overall, TTS, especially the construct consisting of a hairpin DNA (13 pN opening force) and a shearing DNA (20 pN opening force), stands as a valuable tool for the quantification of receptor-transmitted molecular forces within living cells.
Collapse
Affiliation(s)
- Gopal Niraula
- Department
of Physics and Astronomy, Iowa State University, Ames, Iowa 50011, United States
| | - Arghajit Pyne
- Research
Division in Hoxworth Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio 45219, United States
| | - Xuefeng Wang
- Research
Division in Hoxworth Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio 45219, United States
| |
Collapse
|
6
|
Kim DS, Cheah JS, Lai TW, Zhao KX, Foust SR, Julie Lee YR, Lo SH, Heinrich V, Yamada S. Tandem LIM domain-containing proteins, LIMK1 and LMO1, directly bind to force-bearing keratin intermediate filaments. Cell Rep 2024; 43:114480. [PMID: 39003737 DOI: 10.1016/j.celrep.2024.114480] [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: 05/10/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
The cytoskeleton of the cell is constantly exposed to physical forces that regulate cellular functions. Selected members of the LIM (Lin-11, Isl-1, and Mec-3) domain-containing protein family accumulate along force-bearing actin fibers, with evidence supporting that the LIM domain is solely responsible for this force-induced interaction. However, LIM domain's force-induced interactions are not limited to actin. LIMK1 and LMO1, both containing only two tandem LIM domains, are recruited to force-bearing keratin fibers in epithelial cells. This unique recruitment is mediated by their LIM domains and regulated by the sequences outside the LIM domains. Based on in vitro reconstitution of this interaction, LIMK1 and LMO1 directly interact with stretched keratin 8/18 fibers. These results show that LIM domain's mechano-sensing abilities extend to the keratin cytoskeleton, highlighting the diverse role of LIM proteins in force-regulated signaling.
Collapse
Affiliation(s)
- Dah Som Kim
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA
| | - Joleen S Cheah
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA
| | - Tzu Wei Lai
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA
| | - Karen X Zhao
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA
| | - Skylar R Foust
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA
| | - Yuh-Ru Julie Lee
- Department of Plant Biology, University of California, Davis, Davis CA 95616, USA
| | - Su Hao Lo
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis CA 95616 USA
| | - Volkmar Heinrich
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA
| | - Soichiro Yamada
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA.
| |
Collapse
|
7
|
Xie J, Huck WTS, Bao M. Unveiling the Intricate Connection: Cell Volume as a Key Regulator of Mechanotransduction. Annu Rev Biophys 2024; 53:299-317. [PMID: 38424091 DOI: 10.1146/annurev-biophys-030822-035656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The volumes of living cells undergo dynamic changes to maintain the cells' structural and functional integrity in many physiological processes. Minor fluctuations in cell volume can serve as intrinsic signals that play a crucial role in cell fate determination during mechanotransduction. In this review, we discuss the variability of cell volume and its role in vivo, along with an overview of the mechanisms governing cell volume regulation. Additionally, we provide insights into the current approaches used to control cell volume in vitro. Furthermore, we summarize the biological implications of cell volume regulation and discuss recent advances in understanding the fundamental relationship between cell volume and mechanotransduction. Finally, we delve into the potential underlying mechanisms, including intracellular macromolecular crowding and cellular mechanics, that govern the global regulation of cell fate in response to changes in cell volume. By exploring the intricate interplay between cell volume and mechanotransduction, we underscore the importance of considering cell volume as a fundamental signaling cue to unravel the basic principles of mechanotransduction. Additionally, we propose future research directions that can extend our current understanding of cell volume in mechanotransduction. Overall, this review highlights the significance of considering cell volume as a fundamental signal in understanding the basic principles in mechanotransduction and points out the possibility of controlling cell volume to control cell fate, mitigate disease-related damage, and facilitate the healing of damaged tissues.
Collapse
Affiliation(s)
- Jing Xie
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Wilhelm T S Huck
- Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands;
| | - Min Bao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China;
| |
Collapse
|
8
|
Ding S, Chen Y, Huang C, Song L, Liang Z, Wei B. Perception and response of skeleton to mechanical stress. Phys Life Rev 2024; 49:77-94. [PMID: 38564907 DOI: 10.1016/j.plrev.2024.03.011] [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: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
Mechanical stress stands as a fundamental factor in the intricate processes governing the growth, development, morphological shaping, and maintenance of skeletal mass. The profound influence of stress in shaping the skeletal framework prompts the assertion that stress essentially births the skeleton. Despite this acknowledgment, the mechanisms by which the skeleton perceives and responds to mechanical stress remain enigmatic. In this comprehensive review, our scrutiny focuses on the structural composition and characteristics of sclerotin, leading us to posit that it serves as the primary structure within the skeleton responsible for bearing and perceiving mechanical stress. Furthermore, we propose that osteocytes within the sclerotin emerge as the principal mechanical-sensitive cells, finely attuned to perceive mechanical stress. And a detailed analysis was conducted on the possible transmission pathways of mechanical stress from the extracellular matrix to the nucleus.
Collapse
Affiliation(s)
- Sicheng Ding
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Yiren Chen
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Chengshuo Huang
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Lijun Song
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Zhen Liang
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.
| | - Bo Wei
- Department of Minimally invasive spine surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.
| |
Collapse
|
9
|
Mougkogiannis P, Adamatzky A. Thermosensory Spiking Activity of Proteinoid Microspheres Cross-Linked by Actin Filaments. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:12649-12670. [PMID: 38837748 PMCID: PMC11191697 DOI: 10.1021/acs.langmuir.4c01107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/07/2024]
Abstract
Actin, found in all eukaryotic cells as globular (G) or filamentous (F) actin, undergoes polymerization, with G-actin units changing shape to become F-actin. Thermal proteins, or proteinoids, are created by heating amino acids (160-200 °C), forming polymeric chains. These proteinoids can swell in an aqueous solution at around 50 °C, producing hollow microspheres filled with a solution, exhibiting voltage spikes. Our research explores the signaling properties of proteinoids, actin filaments, and hybrid networks combining actin and proteinoids. Proteinoids replicate brain excitation dynamics despite lacking specific membranes or ion channels. We investigate enhancing conductivity and spiking by using pure actin, yielding improved coordination in networks compared with individual filaments or proteinoids. Temperature changes (20 short-peptide supramolecular C to 80 °C) regulate conduction states, demonstrating external control over emergent excitability in protobrain systems. Adding actin to proteinoids reduces spike timing variability, providing a more uniform feature distribution. These findings support theoretical models proposing cytoskeletal matrices for functional specification in synthetic protocell brains, promoting stable interaction complexity. The study concludes that life-like signal encoding can emerge spontaneously within biological polymer scaffolds, incorporating abiotic chemistry.
Collapse
Affiliation(s)
| | - Andrew Adamatzky
- Unconventional Computing
Laboratory, UWE Bristol, Bristol BS16 1QY, U.K.
| |
Collapse
|
10
|
Chinthalapudi K, Heissler SM. Structure, regulation, and mechanisms of nonmuscle myosin-2. Cell Mol Life Sci 2024; 81:263. [PMID: 38878079 PMCID: PMC11335295 DOI: 10.1007/s00018-024-05264-6] [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: 03/11/2024] [Revised: 04/24/2024] [Accepted: 04/30/2024] [Indexed: 06/23/2024]
Abstract
Members of the myosin superfamily of molecular motors are large mechanochemical ATPases that are implicated in an ever-expanding array of cellular functions. This review focuses on mammalian nonmuscle myosin-2 (NM2) paralogs, ubiquitous members of the myosin-2 family of filament-forming motors. Through the conversion of chemical energy into mechanical work, NM2 paralogs remodel and shape cells and tissues. This process is tightly controlled in time and space by numerous synergetic regulation mechanisms to meet cellular demands. We review how recent advances in structural biology together with elegant biophysical and cell biological approaches have contributed to our understanding of the shared and unique mechanisms of NM2 paralogs as they relate to their kinetics, regulation, assembly, and cellular function.
Collapse
Affiliation(s)
- Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Sarah M Heissler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH, 43210, USA.
| |
Collapse
|
11
|
Graziani V, Crosas-Molist E, George SL, Sanz-Moreno V. Organelle adaptations in response to mechanical forces during tumour dissemination. Curr Opin Cell Biol 2024; 88:102345. [PMID: 38479111 DOI: 10.1016/j.ceb.2024.102345] [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: 11/20/2023] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 06/16/2024]
Abstract
Cell migration plays a pivotal role in various biological processes including cancer dissemination and successful metastasis, where the role of mechanical signals is increasingly acknowledged. This review focuses on the intricate mechanisms through which cancer cells modulate their migratory strategies via organelle adaptations in response to the extracellular matrix (ECM). Specifically, the nucleus and mitochondria emerge as pivotal mediators in this process. These organelles serve as sensors, translating mechanical stimuli into rapid metabolic alterations that sustain cell migration. Importantly, prolonged exposure to such stimuli can induce transcriptional or epigenetic changes, ultimately enhancing metastatic traits. Deciphering the intricate interplay between ECM properties and organelle adaptations not only advances our understanding of cytoskeletal dynamics but also holds promise for the development of innovative anti-metastatic therapeutic strategies.
Collapse
Affiliation(s)
- Vittoria Graziani
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London EC1M 6BQ, UK
| | - Eva Crosas-Molist
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London EC1M 6BQ, UK
| | - Samantha L George
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London EC1M 6BQ, UK
| | - Victoria Sanz-Moreno
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
12
|
Tsai FC, Guérin G, Pernier J, Bassereau P. Actin-membrane linkers: Insights from synthetic reconstituted systems. Eur J Cell Biol 2024; 103:151402. [PMID: 38461706 DOI: 10.1016/j.ejcb.2024.151402] [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: 12/03/2023] [Revised: 02/10/2024] [Accepted: 02/28/2024] [Indexed: 03/12/2024] Open
Abstract
At the cell surface, the actin cytoskeleton and the plasma membrane interact reciprocally in a variety of processes related to the remodeling of the cell surface. The actin cytoskeleton has been known to modulate membrane organization and reshape the membrane. To this end, actin-membrane linking molecules play a major role in regulating actin assembly and spatially direct the interaction between the actin cytoskeleton and the membrane. While studies in cells have provided a wealth of knowledge on the molecular composition and interactions of the actin-membrane interface, the complex molecular interactions make it challenging to elucidate the precise actions of the actin-membrane linkers at the interface. Synthetic reconstituted systems, consisting of model membranes and purified proteins, have been a powerful approach to elucidate how actin-membrane linkers direct actin assembly to drive membrane shape changes. In this review, we will focus only on several actin-membrane linkers that have been studied by using reconstitution systems. We will discuss the design principles of these reconstitution systems and how they have contributed to the understanding of the cellular functions of actin-membrane linkers. Finally, we will provide a perspective on future research directions in understanding the intricate actin-membrane interaction.
Collapse
Affiliation(s)
- Feng-Ching Tsai
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Physics of Cells and Cancer, Paris 75005, France.
| | - Gwendal Guérin
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Physics of Cells and Cancer, Paris 75005, France
| | - Julien Pernier
- Tumor Cell Dynamics Unit, Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif 94800, France
| | - Patricia Bassereau
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Physics of Cells and Cancer, Paris 75005, France.
| |
Collapse
|
13
|
Wu W, Zhao Z, Wang Y, Zhu G, Tan K, Liu M, Li L. Biomechanical Effects of Mechanical Stress on Cells Involved in Fracture Healing. Orthop Surg 2024; 16:811-820. [PMID: 38439564 PMCID: PMC10984830 DOI: 10.1111/os.14026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 03/06/2024] Open
Abstract
Fracture healing is a complex staged repair process in which the mechanical environment plays a key role. Bone tissue is very sensitive to mechanical stress stimuli, and the literature suggests that appropriate stress can promote fracture healing by altering cellular function. However, fracture healing is a coupled process involving multiple cell types that balance and limit each other to ensure proper fracture healing. The main cells that function during different stages of fracture healing are different, and the types and molecular mechanisms of stress required are also different. Most previous studies have used a single mechanical stimulus on individual mechanosensitive cells, and there is no relatively uniform standard for the size and frequency of the mechanical stress. Analyzing the mechanisms underlying the effects of mechanical stimulation on the metabolic regulation of signaling pathways in cells such as in bone marrow mesenchymal stem cells (BMSCs), osteoblasts, chondrocytes, and osteoclasts is currently a challenging research hotspot. Grasping how stress affects the function of different cells at the molecular biology level can contribute to the refined management of fracture healing. Therefore, in this review, we summarize the relevant literature and describe the effects of mechanical stress on cells associated with fracture healing, and their possible signaling pathways, for the treatment of fractures and the further development of regenerative medicine.
Collapse
Affiliation(s)
- Weiyong Wu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihui Zhao
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Yongqing Wang
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Gengbao Zhu
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Kemeng Tan
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| | - Meiyue Liu
- Orthopedic Department, The Fourth Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Lili Li
- General Clinical Research Center, Anhui Wanbei Coal-Electricity Group General Hospital, Suzhou, China
| |
Collapse
|
14
|
Nashchekin D, Squires I, Prokop A, St Johnston D. The Shot CH1 domain recognises a distinct form of F-actin during Drosophila oocyte determination. Development 2024; 151:dev202370. [PMID: 38564309 PMCID: PMC11058685 DOI: 10.1242/dev.202370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024]
Abstract
In Drosophila, only one cell in a multicellular female germline cyst is specified as an oocyte and a similar process occurs in mammals. The symmetry-breaking cue for oocyte selection is provided by the fusome, a tubular structure connecting all cells in the cyst. The Drosophila spectraplakin Shot localises to the fusome and translates its asymmetry into a polarised microtubule network that is essential for oocyte specification, but how Shot recognises the fusome is unclear. Here, we demonstrate that the actin-binding domain (ABD) of Shot is necessary and sufficient to localise Shot to the fusome and mediates Shot function in oocyte specification together with the microtubule-binding domains. The calponin homology domain 1 of the Shot ABD recognises fusomal F-actin and requires calponin homology domain 2 to distinguish it from other forms of F-actin in the cyst. By contrast, the ABDs of utrophin, Fimbrin, Filamin, Lifeact and F-tractin do not recognise fusomal F-actin. We therefore propose that Shot propagates fusome asymmetry by recognising a specific conformational state of F-actin on the fusome.
Collapse
Affiliation(s)
- Dmitry Nashchekin
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Iolo Squires
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Andreas Prokop
- The University of Manchester, Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, School of Biology, Manchester M13 9PT, UK
| | - Daniel St Johnston
- The Gurdon Institute and the Department of Genetics, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| |
Collapse
|
15
|
Maeda T, Shimamori K, Kurita H, Tokuraku K, Kuragano M. Amyloid β interferes with wound healing of brain microvascular endothelial cells by disorganizing the actin cytoskeleton. Exp Cell Res 2024; 436:113958. [PMID: 38325585 DOI: 10.1016/j.yexcr.2024.113958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is a disease in which amyloid β (Aβ) is deposited in the cerebral blood vessels, reducing compliance, tearing and weakening of vessel walls, leading to cerebral hemorrhage. The mechanisms by which Aβ leads to focal wall fragmentation and intimal damage are not well understood. We analyzed the motility of human brain microvascular endothelial cells (hBMECs) in real-time using a wound-healing assay. We observed the suppression of cell migration by visualizing Aβ aggregation using quantum dot (QD) nanoprobes. In addition, using QD nanoprobes and a SiR-actin probe, we simultaneously observed Aβ aggregation and F-actin organization in real-time for the first time. Aβ began to aggregate at the edge of endothelial cells, reducing cell motility. In addition, Aβ aggregation disorganized the actin cytoskeleton and induced abnormal actin aggregation. Aβ aggregated actively in the anterior group, where cell motility was active. Our findings may be a first step toward explaining the mechanism by which Aβ causes vascular wall fragility, bleeding, and rebleeding in CAA.
Collapse
Affiliation(s)
- Takuma Maeda
- Graduate School of Engineering, Muroran Institute of Technology, Hokkaido, 050-8585, Japan; Department of Neurosurgery, Ohkawara Neurosurgical Hospital, Hokkaido, 050-0082, Japan; Department of Cerebrovascular Surgery, Saitama Medical University International Medical Center, Saitama, 350-1298, Japan
| | - Keiya Shimamori
- Graduate School of Engineering, Muroran Institute of Technology, Hokkaido, 050-8585, Japan
| | - Hiroki Kurita
- Department of Cerebrovascular Surgery, Saitama Medical University International Medical Center, Saitama, 350-1298, Japan
| | - Kiyotaka Tokuraku
- Graduate School of Engineering, Muroran Institute of Technology, Hokkaido, 050-8585, Japan
| | - Masahiro Kuragano
- Graduate School of Engineering, Muroran Institute of Technology, Hokkaido, 050-8585, Japan.
| |
Collapse
|
16
|
Cao R, Tian H, Tian Y, Fu X. A Hierarchical Mechanotransduction System: From Macro to Micro. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302327. [PMID: 38145330 PMCID: PMC10953595 DOI: 10.1002/advs.202302327] [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: 04/12/2023] [Revised: 10/27/2023] [Indexed: 12/26/2023]
Abstract
Mechanotransduction is a strictly regulated process whereby mechanical stimuli, including mechanical forces and properties, are sensed and translated into biochemical signals. Increasing data demonstrate that mechanotransduction is crucial for regulating macroscopic and microscopic dynamics and functionalities. However, the actions and mechanisms of mechanotransduction across multiple hierarchies, from molecules, subcellular structures, cells, tissues/organs, to the whole-body level, have not been yet comprehensively documented. Herein, the biological roles and operational mechanisms of mechanotransduction from macro to micro are revisited, with a focus on the orchestrations across diverse hierarchies. The implications, applications, and challenges of mechanotransduction in human diseases are also summarized and discussed. Together, this knowledge from a hierarchical perspective has the potential to refresh insights into mechanotransduction regulation and disease pathogenesis and therapy, and ultimately revolutionize the prevention, diagnosis, and treatment of human diseases.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Huimin Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Yan Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Xianghui Fu
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| |
Collapse
|
17
|
Nguyen MT, Ly QK, Kim HJ, Lee W. WAVE2 Is a Vital Regulator in Myogenic Differentiation of Progenitor Cells through the Mechanosensitive MRTFA-SRF Axis. Cells 2023; 13:9. [PMID: 38201213 PMCID: PMC10778525 DOI: 10.3390/cells13010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Skeletal myogenesis is an intricate process involving the differentiation of progenitor cells into myofibers, which is regulated by actin cytoskeletal dynamics and myogenic transcription factors. Although recent studies have demonstrated the pivotal roles of actin-binding proteins (ABPs) as mechanosensors and signal transducers, the biological significance of WAVE2 (Wiskott-Aldrich syndrome protein family member 2), an ABP essential for actin polymerization, in myogenic differentiation of progenitor cells has not been investigated. Our study provides important insights into the regulatory roles played by WAVE2 in the myocardin-related transcription factor A (MRTFA)-serum response factor (SRF) signaling axis and differentiation of myoblasts. We demonstrate that WAVE2 expression is induced during myogenic differentiation and plays a pivotal role in actin cytoskeletal remodeling in C2C12 myoblasts. Knockdown of WAVE2 in C2C12 cells reduced filamentous actin levels, increased globular actin accumulation, and impaired the nuclear translocation of MRTFA. Furthermore, WAVE2 depletion in myoblasts inhibited the expression and transcriptional activity of SRF and suppressed cell proliferation in myoblasts. Consequently, WAVE2 knockdown suppressed myogenic regulatory factors (i.e., MyoD, MyoG, and SMYD1) expressions, thereby hindering the differentiation of myoblasts. Thus, this study suggests that WAVE2 is essential for myogenic differentiation of progenitor cells by modulating the mechanosensitive MRTFA-SRF axis.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (H.-J.K.)
| | - Quoc Kiet Ly
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (H.-J.K.)
| | - Hyun-Jung Kim
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (H.-J.K.)
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (H.-J.K.)
- Channelopathy Research Center, Dongguk University College of Medicine, 32 Dongguk-ro, Goyang 10326, Republic of Korea
| |
Collapse
|
18
|
Wenger J, Brigante A, Fernández Casafuz AB, Bruno L, Monastra A. Inference of the force pattern acting on a semiflexible filament from shape analysis. Phys Rev E 2023; 108:064402. [PMID: 38243502 DOI: 10.1103/physreve.108.064402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/06/2023] [Indexed: 01/21/2024]
Abstract
The study of the active forces acting on semiflexible filaments networks such as the cytoskeleton requires noninvasive tools able to explore the deformation of single filaments in their natural environment. We propose here a practical method based on the solution of the hydrodynamic beam equation in the presence of transverse forces. We found that the derivative of the local curvature presents discontinuities that match the location of the applied forces, in contrast to the smooth curvature function obtained for the case of compressing longitudinal forces. These patterns can be easily appreciated in a kymograph of the curvature, which also reflects the temporal behavior of the forces. We assessed the method performance with numerical simulations describing the deformation of single microtubules provoked by the action of intracellular active forces.
Collapse
Affiliation(s)
- Julieta Wenger
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Física, C1428EGA Buenos Aires, Argentina
| | - Azul Brigante
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Física, C1428EGA Buenos Aires, Argentina
| | - Agustina B Fernández Casafuz
- Universidad de Buenos Aires, Instituto de Cálculo (IC), C1428EGA Buenos Aires, Argentina and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1425FQD Buenos Aires, Argentina
| | - Luciana Bruno
- Universidad de Buenos Aires, Instituto de Cálculo (IC), C1428EGA Buenos Aires, Argentina and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1425FQD Buenos Aires, Argentina
| | - Alejandro Monastra
- Universidad Nacional de General Sarmiento, Instituto de Ciencias, B1613 Los Polvorines, Buenos Aires, Argentina and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1425FQD Buenos Aires, Argentina
| |
Collapse
|
19
|
Komatsu K, Matsuura T, Suzumura T, Ogawa T. Genome-wide transcriptional responses of osteoblasts to different titanium surface topographies. Mater Today Bio 2023; 23:100852. [PMID: 38024842 PMCID: PMC10663851 DOI: 10.1016/j.mtbio.2023.100852] [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: 05/25/2023] [Revised: 10/21/2023] [Accepted: 10/29/2023] [Indexed: 12/01/2023] Open
Abstract
This is the first genome-wide transcriptional profiling study using RNA-sequencing to investigate osteoblast responses to different titanium surface topographies, specifically between machined, smooth and acid-etched, microrough surfaces. Rat femoral osteoblasts were cultured on machine-smooth and acid-etched microrough titanium disks. The culture system was validated through a series of assays confirming reduced osteoblast attachment, slower proliferation, and faster differentiation on microrough surfaces. RNA-sequencing analysis of osteoblasts at an early stage of culture revealed that gene expression was highly correlated (r = 0.975) between the two topographies, but 1.38 % genes were upregulated and 0.37 % were downregulated on microrough surfaces. Upregulated transcripts were enriched for immune system, plasma membrane, response to external stimulus, and positive regulation to stimulus processes. Structural mapping confirmed microrough surface-promoted gene sharing and networking in signaling pathways and immune system/responses. Target-specific pathway analysis revealed that Rho family G-protein signaling pathways and actin genes, responsible for the formation of stress fibers, cytoplasmic projections, and focal adhesion, were upregulated on microrough surfaces without upregulation of core genes triggered by cell-to-cell interactions. Furthermore, disulfide-linked or -targeted extracellular matrix (ECM) or membranous glycoproteins such as laminin, fibronectin, CD36, and thrombospondin were highly expressed on microrough surfaces. Finally, proliferating cell nuclear antigen (PCNA) and cyclin D1, whose co-expression reduces cell proliferation, were upregulated on microrough surfaces. Thus, osteoblasts on microrough surfaces were characterized by upregulation of genes related to a wide range of functions associated with the immune system, stress/stimulus responses, proliferation control, skeletal and cytoplasmic signaling, ECM-integrin receptor interactions, and ECM-membranous glycoprotein interactions, furthering our knowledge of the surface-dependent expression of osteoblastic biomarkers on titanium.
Collapse
Affiliation(s)
- Keiji Komatsu
- Weintraub Center for Reconstructive Biotechnology and the Division of Regenerative and Reconstructive Sciences, UCLA School of Dentistry, Los Angeles, CA, 90095, USA
- Department of Lifetime Oral Health Care Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8549, Japan
| | - Takanori Matsuura
- Weintraub Center for Reconstructive Biotechnology and the Division of Regenerative and Reconstructive Sciences, UCLA School of Dentistry, Los Angeles, CA, 90095, USA
| | - Toshikatsu Suzumura
- Weintraub Center for Reconstructive Biotechnology and the Division of Regenerative and Reconstructive Sciences, UCLA School of Dentistry, Los Angeles, CA, 90095, USA
| | - Takahiro Ogawa
- Weintraub Center for Reconstructive Biotechnology and the Division of Regenerative and Reconstructive Sciences, UCLA School of Dentistry, Los Angeles, CA, 90095, USA
| |
Collapse
|
20
|
Hasannejad F, Montazeri L, Mano JF, Bonakdar S, Fazilat A. Regulation of cell fate by cell imprinting approach in vitro. BIOIMPACTS : BI 2023; 14:29945. [PMID: 38938752 PMCID: PMC11199935 DOI: 10.34172/bi.2023.29945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 09/13/2023] [Accepted: 09/19/2023] [Indexed: 06/29/2024]
Abstract
Cell culture-based technologies are widely utilized in various domains such as drug evaluation, toxicity assessment, vaccine and biopharmaceutical development, reproductive technology, and regenerative medicine. It has been demonstrated that pre-adsorption of extracellular matrix (ECM) proteins including collagen, laminin and fibronectin provide more degrees of support for cell adhesion. The purpose of cell imprinting is to imitate the natural topography of cell membranes by gels or polymers to create a reliable environment for the regulation of cell function. The results of recent studies show that cell imprinting is a tool to guide the behavior of cultured cells by controlling their adhesive interactions with surfaces. Therefore, in this review we aim to compare different cell cultures with the imprinting method and discuss different cell imprinting applications in regenerative medicine, personalized medicine, disease modeling, and cell therapy.
Collapse
Affiliation(s)
- Farkhonde Hasannejad
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Science, Semnan, Iran
- Genetic Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Portugal
| | - Shahin Bonakdar
- National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran
| | - Ahmad Fazilat
- Genetic Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
21
|
Bakhshandeh B, Sorboni SG, Ranjbar N, Deyhimfar R, Abtahi MS, Izady M, Kazemi N, Noori A, Pennisi CP. Mechanotransduction in tissue engineering: Insights into the interaction of stem cells with biomechanical cues. Exp Cell Res 2023; 431:113766. [PMID: 37678504 DOI: 10.1016/j.yexcr.2023.113766] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Stem cells in their natural microenvironment are exposed to biochemical and biophysical cues emerging from the extracellular matrix (ECM) and neighboring cells. In particular, biomechanical forces modulate stem cell behavior, biological fate, and early developmental processes by sensing, interpreting, and responding through a series of biological processes known as mechanotransduction. Local structural changes in the ECM and mechanics are driven by reciprocal activation of the cell and the ECM itself, as the initial deposition of matrix proteins sequentially affects neighboring cells. Recent studies on stem cell mechanoregulation have provided insight into the importance of biomechanical signals on proper tissue regeneration and function and have shown that precise spatiotemporal control of these signals exists in stem cell niches. Against this background, the aim of this work is to review the current understanding of the molecular basis of mechanotransduction by analyzing how biomechanical forces are converted into biological responses via cellular signaling pathways. In addition, this work provides an overview of advanced strategies using stem cells and biomaterial scaffolds that enable precise spatial and temporal control of mechanical signals and offer great potential for the fields of tissue engineering and regenerative medicine will be presented.
Collapse
Affiliation(s)
- Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | | | - Nika Ranjbar
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Roham Deyhimfar
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Maryam Sadat Abtahi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mehrnaz Izady
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Navid Kazemi
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Atefeh Noori
- Department of Biotechnology, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, Denmark.
| |
Collapse
|
22
|
Wang X, Shen Y, Shang M, Liu X, Munn LL. Endothelial mechanobiology in atherosclerosis. Cardiovasc Res 2023; 119:1656-1675. [PMID: 37163659 PMCID: PMC10325702 DOI: 10.1093/cvr/cvad076] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 02/11/2023] [Accepted: 02/21/2023] [Indexed: 05/12/2023] Open
Abstract
Cardiovascular disease (CVD) is a serious health challenge, causing more deaths worldwide than cancer. The vascular endothelium, which forms the inner lining of blood vessels, plays a central role in maintaining vascular integrity and homeostasis and is in direct contact with the blood flow. Research over the past century has shown that mechanical perturbations of the vascular wall contribute to the formation and progression of atherosclerosis. While the straight part of the artery is exposed to sustained laminar flow and physiological high shear stress, flow near branch points or in curved vessels can exhibit 'disturbed' flow. Clinical studies as well as carefully controlled in vitro analyses have confirmed that these regions of disturbed flow, which can include low shear stress, recirculation, oscillation, or lateral flow, are preferential sites of atherosclerotic lesion formation. Because of their critical role in blood flow homeostasis, vascular endothelial cells (ECs) have mechanosensory mechanisms that allow them to react rapidly to changes in mechanical forces, and to execute context-specific adaptive responses to modulate EC functions. This review summarizes the current understanding of endothelial mechanobiology, which can guide the identification of new therapeutic targets to slow or reverse the progression of atherosclerosis.
Collapse
Affiliation(s)
- Xiaoli Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
| | - Yang Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Min Shang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lance L Munn
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
23
|
Deciphering the molecular mechanisms of actin cytoskeleton regulation in cell migration using cryo-EM. Biochem Soc Trans 2023; 51:87-99. [PMID: 36695514 PMCID: PMC9987995 DOI: 10.1042/bst20220221] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/26/2023]
Abstract
The actin cytoskeleton plays a key role in cell migration and cellular morphodynamics in most eukaryotes. The ability of the actin cytoskeleton to assemble and disassemble in a spatiotemporally controlled manner allows it to form higher-order structures, which can generate forces required for a cell to explore and navigate through its environment. It is regulated not only via a complex synergistic and competitive interplay between actin-binding proteins (ABP), but also by filament biochemistry and filament geometry. The lack of structural insights into how geometry and ABPs regulate the actin cytoskeleton limits our understanding of the molecular mechanisms that define actin cytoskeleton remodeling and, in turn, impact emerging cell migration characteristics. With the advent of cryo-electron microscopy (cryo-EM) and advanced computational methods, it is now possible to define these molecular mechanisms involving actin and its interactors at both atomic and ultra-structural levels in vitro and in cellulo. In this review, we will provide an overview of the available cryo-EM methods, applicable to further our understanding of the actin cytoskeleton, specifically in the context of cell migration. We will discuss how these methods have been employed to elucidate ABP- and geometry-defined regulatory mechanisms in initiating, maintaining, and disassembling cellular actin networks in migratory protrusions.
Collapse
|
24
|
Wang J, Yang H, Ma X, Liu J, Li L, Chen L, Wei F. LRP6/filamentous-actin signaling facilitates osteogenic commitment in mechanically induced periodontal ligament stem cells. Cell Mol Biol Lett 2023; 28:7. [PMID: 36694134 PMCID: PMC9872397 DOI: 10.1186/s11658-023-00420-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/12/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Mechanotransduction mechanisms whereby periodontal ligament stem cells (PDLSCs) translate mechanical stress into biochemical signals and thereby trigger osteogenic programs necessary for alveolar bone remodeling are being deciphered. Low-density lipoprotein receptor-related protein 6 (LRP6), a Wnt transmembrane receptor, has been qualified as a key monitor for mechanical cues. However, the role of LRP6 in the mechanotransduction of mechanically induced PDLSCs remains obscure. METHODS The Tension System and tooth movement model were established to determine the expression profile of LRP6. The loss-of-function assay was used to investigate the role of LRP6 on force-regulated osteogenic commitment in PDLSCs. The ability of osteogenic differentiation and proliferation was estimated by alkaline phosphatase (ALP) staining, ALP activity assay, western blotting, quantitative real-time PCR (qRT-PCR), and immunofluorescence. Crystalline violet staining was used to visualize cell morphological change. Western blotting, qRT-PCR, and phalloidin staining were adopted to affirm filamentous actin (F-actin) alteration. YAP nucleoplasmic localization was assessed by immunofluorescence and western blotting. YAP transcriptional response was evaluated by qRT-PCR. Cytochalasin D was used to determine the effects of F-actin on osteogenic commitment and YAP switch behavior in mechanically induced PDLSCs. RESULTS LRP6 was robustly activated in mechanically induced PDLSCs and PDL tissues. LRP6 deficiency impeded force-dependent osteogenic differentiation and proliferation in PDLSCs. Intriguingly, LRP6 loss caused cell morphological aberration, F-actin dynamics disruption, YAP nucleoplasmic relocation, and subsequent YAP inactivation. Moreover, disrupted F-actin dynamics inhibited osteogenic differentiation, proliferation, YAP nuclear translocation, and YAP activation in mechanically induced PDLSCs. CONCLUSIONS We identified that LRP6 in PDLSCs acted as the mechanosensor regulating mechanical stress-inducible osteogenic commitment via the F-actin/YAP cascade. Targeting LRP6 for controlling alveolar bone remodeling may be a prospective therapy to attenuate relapse of orthodontic treatment.
Collapse
Affiliation(s)
- Jixiao Wang
- grid.27255.370000 0004 1761 1174Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan, 250012 Shandong China
| | - Huiqi Yang
- grid.27255.370000 0004 1761 1174Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan, 250012 Shandong China
| | - Xiaobei Ma
- grid.27255.370000 0004 1761 1174Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan, 250012 Shandong China
| | - Jiani Liu
- grid.27255.370000 0004 1761 1174Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan, 250012 Shandong China
| | - Lan Li
- grid.27255.370000 0004 1761 1174Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan, 250012 Shandong China
| | - Lei Chen
- grid.27255.370000 0004 1761 1174Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan, 250012 Shandong China
| | - Fulan Wei
- grid.27255.370000 0004 1761 1174Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan, 250012 Shandong China
| |
Collapse
|
25
|
Sun Q, Pei F, Zhang M, Zhang B, Jin Y, Zhao Z, Wei Q. Curved Nanofiber Network Induces Cellular Bridge Formation to Promote Stem Cell Mechanotransduction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204479. [PMID: 36382560 PMCID: PMC9875655 DOI: 10.1002/advs.202204479] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/26/2022] [Indexed: 06/16/2023]
Abstract
Remarkable exertions are directed to reveal and understand topographic cues that induce cell mechanical sensitive responses including lineage determination. Extracellular matrix (ECM) is the sophisticated ensemble of diverse factors offering the complicated cellular microenvironment to regulate cell behaviors. However, the functions of only a few of these factors are revealed; most of them are still poorly understood. Herein, the focus is on understanding the curved structure in ECM network for regulating stem cell mechanotransduction. A curved nanofiber network mimicking the curved structure in ECM is fabricated by an improved electrospinning technology. Compared with the straight fibers, the curved fibers promote cell bridge formation because of the cytoskeleton tension. The actomyosin filaments are condensed near the curved edge of the non-adhesive bridge in the bridging cells, which generates higher myosin-II-based intracellular force. This force drives cell lineage commitment toward osteogenic differentiation. This study enriches and perfects the knowledge of the effects of topographic cues on cell behaviors and guides the development of novel biomaterials.
Collapse
Affiliation(s)
- Qian Sun
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials and EngineeringSichuan UniversityChengdu610065P. R. China
| | - Fang Pei
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials and EngineeringSichuan UniversityChengdu610065P. R. China
| | - Man Zhang
- College of Biomedical EngineeringSichuan UniversityChengdu610065P. R. China
| | - Bo Zhang
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Ying Jin
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Zhihe Zhao
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Qiang Wei
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials and EngineeringSichuan UniversityChengdu610065P. R. China
- College of Biomedical EngineeringSichuan UniversityChengdu610065P. R. China
| |
Collapse
|
26
|
Ji X, Tian X, Feng S, Zhang L, Wang J, Guo R, Zhu Y, Yu X, Zhang Y, Du H, Zablotskii V, Zhang X. Intermittent F-actin Perturbations by Magnetic Fields Inhibit Breast Cancer Metastasis. RESEARCH (WASHINGTON, D.C.) 2023; 6:0080. [PMID: 36939445 PMCID: PMC10017101 DOI: 10.34133/research.0080] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/31/2023] [Indexed: 02/04/2023]
Abstract
F-actin (filamentous actin) has been shown to be sensitive to mechanical stimuli and play critical roles in cell attachment, migration, and cancer metastasis, but there are very limited ways to perturb F-actin dynamics with low cell toxicity. Magnetic field is a noninvasive and reversible physical tool that can easily penetrate cells and human bodies. Here, we show that 0.1/0.4-T 4.2-Hz moderate-intensity low-frequency rotating magnetic field-induced electric field could directly decrease F-actin formation in vitro and in vivo, which results in decreased breast cancer cell migration, invasion, and attachment. Moreover, low-frequency rotating magnetic fields generated significantly different effects on F-actin in breast cancer vs. noncancerous cells, including F-actin number and their recovery after magnetic field retrieval. Using an intermittent treatment modality, low-frequency rotating magnetic fields could significantly reduce mouse breast cancer metastasis, prolong mouse survival by 31.5 to 46.0% (P < 0.0001), and improve their overall physical condition. Therefore, our work demonstrates that low-frequency rotating magnetic fields not only can be used as a research tool to perturb F-actin but also can inhibit breast cancer metastasis through F-actin modulation while having minimum effects on normal cells, which reveals their potential to be developed as temporal-controlled, noninvasive, and high-penetration physical treatments for metastatic cancer.
Collapse
Affiliation(s)
- Xinmiao Ji
- High Magnetic Field Laboratory of CAS (CHMFL), CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology,
HFIPS, Hefei, Anhui 230031, P.R China
| | - Xiaofei Tian
- Institutes of Physical Science and Information Technology,
Anhui University, Hefei, Anhui, 230601, P. R. China
| | - Shuang Feng
- High Magnetic Field Laboratory of CAS (CHMFL), CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology,
HFIPS, Hefei, Anhui 230031, P.R China
| | - Lei Zhang
- High Magnetic Field Laboratory of CAS (CHMFL), CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology,
HFIPS, Hefei, Anhui 230031, P.R China
| | - Junjun Wang
- High Magnetic Field Laboratory of CAS (CHMFL), CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology,
HFIPS, Hefei, Anhui 230031, P.R China
| | - Ruowen Guo
- High Magnetic Field Laboratory of CAS (CHMFL), CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology,
HFIPS, Hefei, Anhui 230031, P.R China
- Science Island Branch of Graduate School,
University of Science and Technology of China, Hefei, Anhui 230031, P.R China
| | - Yiming Zhu
- High Magnetic Field Laboratory of CAS (CHMFL), CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology,
HFIPS, Hefei, Anhui 230031, P.R China
- Science Island Branch of Graduate School,
University of Science and Technology of China, Hefei, Anhui 230031, P.R China
| | - Xin Yu
- High Magnetic Field Laboratory of CAS (CHMFL), CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology,
HFIPS, Hefei, Anhui 230031, P.R China
- Science Island Branch of Graduate School,
University of Science and Technology of China, Hefei, Anhui 230031, P.R China
| | - Yongsen Zhang
- High Magnetic Field Laboratory of CAS (CHMFL), CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology,
HFIPS, Hefei, Anhui 230031, P.R China
| | - Haifeng Du
- High Magnetic Field Laboratory of CAS (CHMFL), CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology,
HFIPS, Hefei, Anhui 230031, P.R China
| | - Vitalii Zablotskii
- Institute of Physics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Xin Zhang
- High Magnetic Field Laboratory of CAS (CHMFL), CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology,
HFIPS, Hefei, Anhui 230031, P.R China
- Institutes of Physical Science and Information Technology,
Anhui University, Hefei, Anhui, 230601, P. R. China
- Science Island Branch of Graduate School,
University of Science and Technology of China, Hefei, Anhui 230031, P.R China
- International Magnetobiology Frontier Research Center, Science Island, Hefei 230031, P.R. China
- Address correspondence to:
| |
Collapse
|
27
|
Single-molecule characterization of subtype-specific β1 integrin mechanics. Nat Commun 2022; 13:7471. [PMID: 36463259 PMCID: PMC9719539 DOI: 10.1038/s41467-022-35173-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
Although integrins are known to be mechanosensitive and to possess many subtypes that have distinct physiological roles, single molecule studies of force exertion have thus far been limited to RGD-binding integrins. Here, we show that integrin α4β1 and RGD-binding integrins (αVβ1 and α5β1) require markedly different tension thresholds to support cell spreading. Furthermore, actin assembled downstream of α4β1 forms cross-linked networks in circularly spread cells, is in rapid retrograde flow, and exerts low forces from actin polymerization. In contrast, actin assembled downstream of αVβ1 forms stress fibers linking focal adhesions in elongated cells, is in slow retrograde flow, and matures to exert high forces (>54-pN) via myosin II. Conformational activation of both integrins occurs below 12-pN, suggesting that post-activation subtype-specific cytoskeletal remodeling imposes the higher threshold for spreading on RGD substrates. Multiple layers of single integrin mechanics for activation, mechanotransduction and cytoskeleton remodeling revealed here may underlie subtype-dependence of diverse processes such as somite formation and durotaxis.
Collapse
|
28
|
Wu G, Yang C, Bruce HL, Roy BC, Li X, Zhang C. Effects of Alternating Electric Field Assisted Freezing-Thawing-Aging Sequence on Data-Independent Acquisition Quantitative Proteomics of Longissimus dorsi Muscle. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:12990-13001. [PMID: 36166831 DOI: 10.1021/acs.jafc.2c04207] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
This study was designed to investigate the differences in the proteomes of bovine Longissimus dorsi (LD) muscle during an alternating electric field (AEF)-assisted freezing-thawing-aging sequence based on a data-independent acquisition strategy. When compared to that of the only postmortem aging (OA) group, the meat quality of the freezing-thawing-aging sequence (FA) and AEF-assisted freezing-thawing-aging sequence (EA) groups showed a declining trend. However, the group assisted by AEF was significantly enhanced in color, water-holding capacity, and tenderness. Three hundred fifty-two proteins in LD muscle were differentially abundant proteins (DAPs) among FA, EA, and OA treatments. Furthermore, among the 40 DAPs in the FA versus EA comparison, 5 DAPs with variable importance in projection scores higher than 1 were identified as biochemical markers of beef quality. Bioinformatic analysis revealed that most of these proteins were involved in structural constituents of ribosome and catalytic activity. These results provide a basis for further understanding the quality of beef following a freezing-thawing-aging sequence assisted by AEF.
Collapse
Affiliation(s)
- Guangyu Wu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-products Processing, Ministry of Agriculture and Rural Affairs, Beijing100193, P.R. China
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AlbertaT6G 2P5, Canada
| | - Chuan Yang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-products Processing, Ministry of Agriculture and Rural Affairs, Beijing100193, P.R. China
| | - Heather L Bruce
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AlbertaT6G 2P5, Canada
| | - Bimol C Roy
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AlbertaT6G 2P5, Canada
| | - Xia Li
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-products Processing, Ministry of Agriculture and Rural Affairs, Beijing100193, P.R. China
| | - Chunhui Zhang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-products Processing, Ministry of Agriculture and Rural Affairs, Beijing100193, P.R. China
| |
Collapse
|
29
|
Tang C, Wang X, D'Urso M, van der Putten C, Kurniawan NA. 3D Interfacial and Spatiotemporal Regulation of Human Neuroepithelial Organoids. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201106. [PMID: 35667878 PMCID: PMC9353482 DOI: 10.1002/advs.202201106] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/20/2022] [Indexed: 06/15/2023]
Abstract
Neuroepithelial (NE) organoids with dorsal-ventral patterning provide a useful three-dimensional (3D) in vitro model to interrogate neural tube formation during early development of the central nervous system. Understanding the fundamental processes behind the cellular self-organization in NE organoids holds the key to the engineering of organoids with higher, more in vivo-like complexity. However, little is known about the cellular regulation driving the NE development, especially in the presence of interfacial cues from the microenvironment. Here a simple 3D culture system that allows generation and manipulation of NE organoids from human-induced pluripotent stem cells (hiPSCs), displaying developmental phases of hiPSC differentiation and self-aggregation, first into NE cysts with lumen structure and then toward NE organoids with floor-plate patterning, is established. Longitudinal inhibition reveals distinct and dynamic roles of actomyosin contractility and yes-associated protein (YAP) signaling in governing these phases. By growing NE organoids on culture chips containing anisotropic surfaces or confining microniches, it is further demonstrated that interfacial cues can sensitively exert dimension-dependent influence on luminal cyst and organoid morphology, successful floor-plate patterning, as well as cytoskeletal regulation and YAP activity. This study therefore sheds new light on how organoid and tissue architecture can be steered through intracellular and extracellular means.
Collapse
Affiliation(s)
- Chunling Tang
- Department of Biomedical EngineeringEindhoven University of TechnologyPO Box 513Eindhoven5600 MBThe Netherlands
- Institute for Complex Molecular SystemsPO Box 513Eindhoven5600 MBThe Netherlands
| | - Xinhui Wang
- Department of Biomedical EngineeringEindhoven University of TechnologyPO Box 513Eindhoven5600 MBThe Netherlands
- Institute for Complex Molecular SystemsPO Box 513Eindhoven5600 MBThe Netherlands
| | - Mirko D'Urso
- Department of Biomedical EngineeringEindhoven University of TechnologyPO Box 513Eindhoven5600 MBThe Netherlands
- Institute for Complex Molecular SystemsPO Box 513Eindhoven5600 MBThe Netherlands
| | - Cas van der Putten
- Department of Biomedical EngineeringEindhoven University of TechnologyPO Box 513Eindhoven5600 MBThe Netherlands
- Institute for Complex Molecular SystemsPO Box 513Eindhoven5600 MBThe Netherlands
| | - Nicholas A. Kurniawan
- Department of Biomedical EngineeringEindhoven University of TechnologyPO Box 513Eindhoven5600 MBThe Netherlands
- Institute for Complex Molecular SystemsPO Box 513Eindhoven5600 MBThe Netherlands
| |
Collapse
|
30
|
Jasnin M, Hervy J, Balor S, Bouissou A, Proag A, Voituriez R, Schneider J, Mangeat T, Maridonneau-Parini I, Baumeister W, Dmitrieff S, Poincloux R. Elasticity of podosome actin networks produces nanonewton protrusive forces. Nat Commun 2022; 13:3842. [PMID: 35789161 PMCID: PMC9253342 DOI: 10.1038/s41467-022-30652-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/10/2022] [Indexed: 11/26/2022] Open
Abstract
Actin filaments assemble into force-generating systems involved in diverse cellular functions, including cell motility, adhesion, contractility and division. It remains unclear how networks of actin filaments, which individually generate piconewton forces, can produce forces reaching tens of nanonewtons. Here we use in situ cryo-electron tomography to unveil how the nanoscale architecture of macrophage podosomes enables basal membrane protrusion. We show that the sum of the actin polymerization forces at the membrane is not sufficient to explain podosome protrusive forces. Quantitative analysis of podosome organization demonstrates that the core is composed of a dense network of bent actin filaments storing elastic energy. Theoretical modelling of the network as a spring-loaded elastic material reveals that it exerts forces of a few tens of nanonewtons, in a range similar to that evaluated experimentally. Thus, taking into account not only the interface with the membrane but also the bulk of the network, is crucial to understand force generation by actin machineries. Our integrative approach sheds light on the elastic behavior of dense actin networks and opens new avenues to understand force production inside cells.
Collapse
Affiliation(s)
- Marion Jasnin
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried, Germany.
| | - Jordan Hervy
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| | - Stéphanie Balor
- Plateforme de Microscopie Électronique Intégrative, Centre de Biologie Intégrative, CNRS, UPS, Toulouse, France
| | - Anaïs Bouissou
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Amsha Proag
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Jonathan Schneider
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Thomas Mangeat
- LITC Core Facility, Centre de Biologie Integrative, Université de Toulouse, CNRS, UPS, 31062, Toulouse, France
| | | | - Wolfgang Baumeister
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Serge Dmitrieff
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France.
| | - Renaud Poincloux
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
31
|
Jia H, Flommersfeld J, Heymann M, Vogel SK, Franquelim HG, Brückner DB, Eto H, Broedersz CP, Schwille P. 3D printed protein-based robotic structures actuated by molecular motor assemblies. NATURE MATERIALS 2022; 21:703-709. [PMID: 35618822 PMCID: PMC9156402 DOI: 10.1038/s41563-022-01258-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 04/13/2022] [Indexed: 06/10/2023]
Abstract
Upscaling motor protein activity to perform work in man-made devices has long been an ambitious goal in bionanotechnology. The use of hierarchical motor assemblies, as realized in sarcomeres, has so far been complicated by the challenges of arranging sufficiently high numbers of motor proteins with nanoscopic precision. Here, we describe an alternative approach based on actomyosin cortex-like force production, allowing low complexity motor arrangements in a contractile meshwork that can be coated onto soft objects and locally activated by ATP. The design is reminiscent of a motorized exoskeleton actuating protein-based robotic structures from the outside. It readily supports the connection and assembly of micro-three-dimensional printed modules into larger structures, thereby scaling up mechanical work. We provide an analytical model of force production in these systems and demonstrate the design flexibility by three-dimensional printed units performing complex mechanical tasks, such as microhands and microarms that can grasp and wave following light activation.
Collapse
Affiliation(s)
- Haiyang Jia
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Johannes Flommersfeld
- Arnold Sommerfeld Center for Theoretical Physics, Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Michael Heymann
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany
| | - Sven K Vogel
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | - David B Brückner
- Arnold Sommerfeld Center for Theoretical Physics, Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hiromune Eto
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Chase P Broedersz
- Arnold Sommerfeld Center for Theoretical Physics, Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany.
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| | - Petra Schwille
- Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
32
|
Yang Y, Chu C, Xiao W, Liu L, Man Y, Lin J, Qu Y. Strategies for advanced particulate bone substitutes regulating the osteo-immune microenvironment. Biomed Mater 2022; 17. [PMID: 35168224 DOI: 10.1088/1748-605x/ac5572] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/15/2022] [Indexed: 02/05/2023]
Abstract
The usage of bone substitute granule materials has improved the clinical results of alveolar bone deficiencies treatment and thus broadened applications in implant dentistry. However, because of the complicated mechanisms controlling the foreign body response, no perfect solution can avoid the fibrotic encapsulation of materials till now, which may impair the results of bone regeneration, even cause the implant materials rejection. Recently, the concept of 'osteoimmunology' has been stressed. The outcomes of bone regeneration are proved to be related to the bio-physicochemical properties of biomaterials, which allow them to regulate the biological behaviours of both innate and adaptive immune cells. With the development of single cell transcriptome, the truly heterogeneity of osteo-immune cells has been clarifying, which is helpful to overcome the limitations of traditional M1/M2 macrophage nomenclature and drive the advancements of particulate biomaterials applications. This review aims at introducing the mechanisms of optimal osseointegration regulated by immune systems and provides feasible strategies for the design of next generation 'osteoimmune-smart' particulate bone substitute materials in dental clinic.
Collapse
Affiliation(s)
- Yang Yang
- Department of Oral Implantology & Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Chenyu Chu
- Department of Oral Implantology & Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Wenlan Xiao
- Department of Oral Implantology & Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Li Liu
- State Key Laboratory of Biotherapy and Laboratory, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yi Man
- Department of Oral Implantology & Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Jie Lin
- Department of Oral Implantology & Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yili Qu
- Department of Oral Implantology & Department of Prosthodontics & State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People's Republic of China
| |
Collapse
|
33
|
Abstract
Single-molecule magnetic tweezers deliver magnetic force and torque to single target molecules, permitting the study of dynamic changes in biomolecular structures and their interactions. Because the magnetic tweezer setups can generate magnetic fields that vary slowly over tens of millimeters-far larger than the nanometer scale of the single molecule events being observed-this technique can maintain essentially constant force levels during biochemical experiments while generating a biologically meaningful force on the order of 1-100 pN. When using bead-tether constructs to pull on single molecules, smaller magnetic beads and shorter submicrometer tethers improve dynamic response times and measurement precision. In addition, employing high-speed cameras, stronger light sources, and a graphics programming unit permits true high-resolution single-molecule magnetic tweezers that can track nanometer changes in target molecules on a millisecond or even submillisecond time scale. The unique force-clamping capacity of the magnetic tweezer technique provides a way to conduct measurements under near-equilibrium conditions and directly map the energy landscapes underlying various molecular phenomena. High-resolution single-molecule magnetic tweezers can thus be used to monitor crucial conformational changes in single-protein molecules, including those involved in mechanotransduction and protein folding. Expected final online publication date for the Annual Review of Biochemistry, Volume 91 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Hyun-Kyu Choi
- Wallace H. Coulter Department of Biomedical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Hyun Gyu Kim
- School of Biological Sciences and Institute for Molecular Biology and Genetics, Seoul National University, Seoul, South Korea;
| | - Min Ju Shon
- Department of Physics and School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science & Technology (POSTECH), Pohang, South Korea;
| | - Tae-Young Yoon
- School of Biological Sciences and Institute for Molecular Biology and Genetics, Seoul National University, Seoul, South Korea;
| |
Collapse
|
34
|
Keen AN, Payne LA, Mehta V, Rice A, Simpson LJ, Pang KL, del Rio Hernandez A, Reader JS, Tzima E. Eukaryotic initiation factor 6 regulates mechanical responses in endothelial cells. J Cell Biol 2022; 221:e202005213. [PMID: 35024764 PMCID: PMC8763864 DOI: 10.1083/jcb.202005213] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/11/2021] [Accepted: 12/08/2021] [Indexed: 12/22/2022] Open
Abstract
The repertoire of extratranslational functions of components of the protein synthesis apparatus is expanding to include control of key cell signaling networks. However, very little is known about noncanonical functions of members of the protein synthesis machinery in regulating cellular mechanics. We demonstrate that the eukaryotic initiation factor 6 (eIF6) modulates cellular mechanobiology. eIF6-depleted endothelial cells, under basal conditions, exhibit unchanged nascent protein synthesis, polysome profiles, and cytoskeleton protein expression, with minimal effects on ribosomal biogenesis. In contrast, using traction force and atomic force microscopy, we show that loss of eIF6 leads to reduced stiffness and force generation accompanied by cytoskeletal and focal adhesion defects. Mechanistically, we show that eIF6 is required for the correct spatial mechanoactivation of ERK1/2 via stabilization of an eIF6-RACK1-ERK1/2-FAK mechanocomplex, which is necessary for force-induced remodeling. These results reveal an extratranslational function for eIF6 and a novel paradigm for how mechanotransduction, the cellular cytoskeleton, and protein translation constituents are linked.
Collapse
Affiliation(s)
- Adam N. Keen
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Luke A. Payne
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Vedanta Mehta
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK
| | - Lisa J. Simpson
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kar Lai Pang
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Armando del Rio Hernandez
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK
| | - John S. Reader
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ellie Tzima
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
35
|
Qu C, Roth R, Puapatanakul P, Loitman C, Hammad D, Genin GM, Miner JH, Suleiman HY. Three-Dimensional Visualization of the Podocyte Actin Network Using Integrated Membrane Extraction, Electron Microscopy, and Machine Learning. J Am Soc Nephrol 2022; 33:155-173. [PMID: 34758982 PMCID: PMC8763187 DOI: 10.1681/asn.2021020182] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 10/19/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Actin stress fibers are abundant in cultured cells, but little is known about them in vivo. In podocytes, much evidence suggests that mechanobiologic mechanisms underlie podocyte shape and adhesion in health and in injury, with structural changes to actin stress fibers potentially responsible for pathologic changes to cell morphology. However, this hypothesis is difficult to rigorously test in vivo due to challenges with visualization. A technology to image the actin cytoskeleton at high resolution is needed to better understand the role of structures such as actin stress fibers in podocytes. METHODS We developed the first visualization technique capable of resolving the three-dimensional cytoskeletal network in mouse podocytes in detail, while definitively identifying the proteins that comprise this network. This technique integrates membrane extraction, focused ion-beam scanning electron microscopy, and machine learning image segmentation. RESULTS Using isolated mouse glomeruli from healthy animals, we observed actin cables and intermediate filaments linking the interdigitated podocyte foot processes to newly described contractile actin structures, located at the periphery of the podocyte cell body. Actin cables within foot processes formed a continuous, mesh-like, electron-dense sheet that incorporated the slit diaphragms. CONCLUSIONS Our new technique revealed, for the first time, the detailed three-dimensional organization of actin networks in healthy podocytes. In addition to being consistent with the gel compression hypothesis, which posits that foot processes connected by slit diaphragms act together to counterbalance the hydrodynamic forces across the glomerular filtration barrier, our data provide insight into how podocytes respond to mechanical cues from their surrounding environment.
Collapse
Affiliation(s)
- Chengqing Qu
- Department of Mechanical Engineering, National Science Foundation Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, Missouri
| | - Robyn Roth
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Charles Loitman
- Division of Nephrology, Washington University School of Medicine, St. Louis, Missouri
| | - Dina Hammad
- Division of Nephrology, Washington University School of Medicine, St. Louis, Missouri
| | - Guy M. Genin
- Department of Mechanical Engineering, National Science Foundation Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, Missouri
| | - Jeffrey H. Miner
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri,Division of Nephrology, Washington University School of Medicine, St. Louis, Missouri
| | - Hani Y. Suleiman
- Division of Nephrology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
36
|
Song M, Kwon S. Enhanced Cellular Permeation Efficiency Through Mechanical Vibration-induced Actin Cytoskeleton Changes in Human Nasal Epithelial Cells. BIOTECHNOL BIOPROC E 2021. [DOI: 10.1007/s12257-021-0070-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
37
|
Shen Y, Zhang W, Li G, Ning P, Li Z, Chen H, Wei X, Pan X, Qin Y, He B, Yu Z, Cheng Y. Adaptive Control of Nanomotor Swarms for Magnetic-Field-Programmed Cancer Cell Destruction. ACS NANO 2021; 15:20020-20031. [PMID: 34807565 DOI: 10.1021/acsnano.1c07615] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Magnetic nanomotors (MNMs), powered by a magnetic field, are ideal platforms to achieve versatile biomedical applications in a collective and spatiotemporal fashion. Although the programmable swarm of MNMs that mimics the highly ordered behaviors of living creatures has been extensively studied at the microscale, it is of vital importance to manipulate MNM swarms at the nanoscale for on-demand tasks at the cellular level. In this work, a Cy5-tagged caspase-3-specific peptide-modified MNM is designed, and the adaptive control behaviors of MNM swarms are revealed in lysosomes to induce the cancer cell apoptosis under a rotating magnetic field (RMF). A magneto-programmed vortex is predicted to occur with swarms under RMF by the finite element method model and verified in vitro. According to the dynamic model and numerical simulation, the critical rotating frequency under which MNMs are out of step is strongly correlated to their assembling and swarming properties. The adaptivity of swarms maximizes the synchronous rotation to achieve an optimal energy conversion rate. The frequency-adapted controllability of MNM swarms for cancer cell apoptosis is observed in real time in vitro and in vivo. This work provides theoretical and experimental insights to adaptively control MNM swarms for cancer treatment.
Collapse
Affiliation(s)
- Yajing Shen
- Shanghai East Hospital, School of Medicine, Tongji University, 1800 Yuntai Road, Shanghai 200120, China
| | - Wei Zhang
- College of Electronics and Information Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Gang Li
- College of Electronics and Information Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Peng Ning
- Shanghai East Hospital, School of Medicine, Tongji University, 1800 Yuntai Road, Shanghai 200120, China
| | - Zhenguang Li
- Shanghai East Hospital, School of Medicine, Tongji University, 1800 Yuntai Road, Shanghai 200120, China
| | - Haotian Chen
- Shanghai East Hospital, School of Medicine, Tongji University, 1800 Yuntai Road, Shanghai 200120, China
| | - Xueyan Wei
- Shanghai East Hospital, School of Medicine, Tongji University, 1800 Yuntai Road, Shanghai 200120, China
| | - Xin Pan
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 1800 Yuntai Road, Shanghai 200120, China
| | - Yao Qin
- Shanghai East Hospital, School of Medicine, Tongji University, 1800 Yuntai Road, Shanghai 200120, China
| | - Bin He
- College of Electronics and Information Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Zuoren Yu
- Shanghai East Hospital, School of Medicine, Tongji University, 1800 Yuntai Road, Shanghai 200120, China
| | - Yu Cheng
- Shanghai East Hospital, School of Medicine, Tongji University, 1800 Yuntai Road, Shanghai 200120, China
| |
Collapse
|
38
|
Kennedy Z, Newberg J, Goelzer M, Judex S, Fitzpatrick CK, Uzer G. Modeling stem cell nucleus mechanics using confocal microscopy. Biomech Model Mechanobiol 2021; 20:2361-2372. [PMID: 34424419 PMCID: PMC8599651 DOI: 10.1007/s10237-021-01513-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 08/14/2021] [Indexed: 11/24/2022]
Abstract
Nuclear mechanics is emerging as a key component of stem cell function and differentiation. While changes in nuclear structure can be visually imaged with confocal microscopy, mechanical characterization of the nucleus and its sub-cellular components require specialized testing equipment. A computational model permitting cell-specific mechanical information directly from confocal and atomic force microscopy of cell nuclei would be of great value. Here, we developed a computational framework for generating finite element models of isolated cell nuclei from multiple confocal microscopy scans and simple atomic force microscopy (AFM) tests. Confocal imaging stacks of isolated mesenchymal stem cells were converted into finite element models and siRNA-mediated Lamin A/C depletion isolated chromatin and Lamin A/C structures. Using AFM-measured experimental stiffness values, a set of conversion factors were determined for both chromatin and Lamin A/C to map the voxel intensity of the original images to the element stiffness, allowing the prediction of nuclear stiffness in an additional set of other nuclei. The developed computational framework will identify the contribution of a multitude of sub-nuclear structures and predict global nuclear stiffness of multiple nuclei based on simple nuclear isolation protocols, confocal images and AFM tests.
Collapse
Affiliation(s)
- Zeke Kennedy
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Drive, MSd-2085, Boise, ID, 83725-2085, USA
| | - Joshua Newberg
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Drive, MSd-2085, Boise, ID, 83725-2085, USA
| | - Matthew Goelzer
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Drive, MSd-2085, Boise, ID, 83725-2085, USA
| | - Stefan Judex
- Biomedical Engineering, Stony Brook University, Stony Brook, USA
| | - Clare K Fitzpatrick
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Drive, MSd-2085, Boise, ID, 83725-2085, USA
| | - Gunes Uzer
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Drive, MSd-2085, Boise, ID, 83725-2085, USA.
| |
Collapse
|
39
|
El-Rashidy AA, El Moshy S, Radwan IA, Rady D, Abbass MMS, Dörfer CE, Fawzy El-Sayed KM. Effect of Polymeric Matrix Stiffness on Osteogenic Differentiation of Mesenchymal Stem/Progenitor Cells: Concise Review. Polymers (Basel) 2021; 13:2950. [PMID: 34502988 PMCID: PMC8434088 DOI: 10.3390/polym13172950] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 01/23/2023] Open
Abstract
Mesenchymal stem/progenitor cells (MSCs) have a multi-differentiation potential into specialized cell types, with remarkable regenerative and therapeutic results. Several factors could trigger the differentiation of MSCs into specific lineages, among them the biophysical and chemical characteristics of the extracellular matrix (ECM), including its stiffness, composition, topography, and mechanical properties. MSCs can sense and assess the stiffness of extracellular substrates through the process of mechanotransduction. Through this process, the extracellular matrix can govern and direct MSCs' lineage commitment through complex intracellular pathways. Hence, various biomimetic natural and synthetic polymeric matrices of tunable stiffness were developed and further investigated to mimic the MSCs' native tissues. Customizing scaffold materials to mimic cells' natural environment is of utmost importance during the process of tissue engineering. This review aims to highlight the regulatory role of matrix stiffness in directing the osteogenic differentiation of MSCs, addressing how MSCs sense and respond to their ECM, in addition to listing different polymeric biomaterials and methods used to alter their stiffness to dictate MSCs' differentiation towards the osteogenic lineage.
Collapse
Affiliation(s)
- Aiah A. El-Rashidy
- Biomaterials Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt;
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
| | - Sara El Moshy
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Israa Ahmed Radwan
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Dina Rady
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Marwa M. S. Abbass
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| | - Christof E. Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24105 Kiel, Germany;
| | - Karim M. Fawzy El-Sayed
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt; (S.E.M.); (I.A.R.); (D.R.); (M.M.S.A.)
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24105 Kiel, Germany;
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
40
|
Fragoulis A, Biller K, Fragoulis S, Lex D, Uhlig S, Reiss LK. Reference Gene Selection for Gene Expression Analyses in Mouse Models of Acute Lung Injury. Int J Mol Sci 2021; 22:ijms22157853. [PMID: 34360619 PMCID: PMC8346155 DOI: 10.3390/ijms22157853] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/12/2021] [Accepted: 07/19/2021] [Indexed: 12/21/2022] Open
Abstract
qRT-PCR still remains the most widely used method for quantifying gene expression levels, although newer technologies such as next generation sequencing are becoming increasingly popular. A critical, yet often underappreciated, problem when analysing qRT-PCR data is the selection of suitable reference genes. This problem is compounded in situations where up to 25% of all genes may change (e.g., due to leukocyte invasion), as is typically the case in ARDS. Here, we examined 11 widely used reference genes for their suitability in commonly used models of acute lung injury (ALI): ventilator-induced lung injury (VILI), in vivo and ex vivo, lipopolysaccharide plus mechanical ventilation (MV), and hydrochloric acid plus MV. The stability of reference gene expression was determined using the NormFinder, BestKeeper, and geNorm algorithms. We then proceeded with the geNorm results because this is the only algorithm that provides the number of reference genes required to achieve normalisation. We chose interleukin-6 (Il-6) and C-X-C motif ligand 1 (Cxcl-1) as the genes of interest to analyse and demonstrate the impact of inappropriate normalisation. Reference gene stability differed between the ALI models and even within the subgroup of VILI models, no common reference gene index (RGI) could be determined. NormFinder, BestKeeper, and geNorm produced slightly different, but comparable results. Inappropriate normalisation of Il-6 and Cxcl1 gene expression resulted in significant misinterpretation in all four ALI settings. In conclusion, choosing an inappropriate normalisation strategy can introduce different kinds of bias such as gain or loss as well as under- or overestimation of effects, affecting the interpretation of gene expression data.
Collapse
Affiliation(s)
- Athanassios Fragoulis
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen University, 52074 Aachen, Germany;
| | - Kristina Biller
- Department of Pharmacology and Toxicology, Uniklinik RWTH Aachen University, 52074 Aachen, Germany; (K.B.); (S.F.); (D.L.); (S.U.)
| | - Stephanie Fragoulis
- Department of Pharmacology and Toxicology, Uniklinik RWTH Aachen University, 52074 Aachen, Germany; (K.B.); (S.F.); (D.L.); (S.U.)
| | - Dennis Lex
- Department of Pharmacology and Toxicology, Uniklinik RWTH Aachen University, 52074 Aachen, Germany; (K.B.); (S.F.); (D.L.); (S.U.)
| | - Stefan Uhlig
- Department of Pharmacology and Toxicology, Uniklinik RWTH Aachen University, 52074 Aachen, Germany; (K.B.); (S.F.); (D.L.); (S.U.)
| | - Lucy Kathleen Reiss
- Department of Pharmacology and Toxicology, Uniklinik RWTH Aachen University, 52074 Aachen, Germany; (K.B.); (S.F.); (D.L.); (S.U.)
- Correspondence:
| |
Collapse
|
41
|
Zhao X, Ou G, Lei M, Zhang Y, Li L, Ge A, Wang Y, Li Y, Liu BF. Rapid generation of hybrid biochemical/mechanical cues in heterogeneous droplets for high-throughput screening of cellular responses. LAB ON A CHIP 2021; 21:2691-2701. [PMID: 34165109 DOI: 10.1039/d1lc00209k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Cells in their native microenvironment are subjected to varying combinations of biochemical cues and mechanical cues in a wide range. Although many signaling pathways have been found to be responsive for extracellular cues, little is known about how biochemical cues crosstalk with mechanical cues in a complex microenvironment. Here, we introduced heterogeneous droplets on a microchip, which were rapidly assembled by combining wettability-patterned microchip and programmed droplet manipulations, for a high-throughput cell screening of the varying combinations of biochemical cues and mechanical cues. This platform constructed a heterogeneous droplet/microgel array with orthogonal gradual chemicals and materials, which was further applied to analyze the cellular Wnt/β-catenin signaling in response to varying combinations of Wnt ligands and substrate stiffness. Thus, this device provides a powerful multiplexed bioassay platform for drug development, tissue engineering, and stem cell screening.
Collapse
Affiliation(s)
- Xing Zhao
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Gaozhi Ou
- School of Sports, China University of Geosciences, Wuhan, 430074, China
| | - Mengcheng Lei
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Yang Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518100, China
| | - Lina Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Anle Ge
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Yachao Wang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
42
|
Li H, Zhang C, Hu Y, Liu P, Sun F, Chen W, Zhang X, Ma J, Wang W, Wang L, Wu P, Liu Z. A reversible shearing DNA probe for visualizing mechanically strong receptors in living cells. Nat Cell Biol 2021; 23:642-651. [PMID: 34059812 DOI: 10.1038/s41556-021-00691-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 04/28/2021] [Indexed: 02/05/2023]
Abstract
In the last decade, DNA-based tension sensors have made significant contributions to the study of the importance of mechanical forces in many biological systems. Albeit successful, one shortcoming of these techniques is their inability to reversibly measure receptor forces in a higher regime (that is, >20 pN), which limits our understanding of the molecular details of mechanochemical transduction in living cells. Here, we developed a reversible shearing DNA-based tension probe (RSDTP) for probing molecular piconewton-scale forces between 4 and 60 pN transmitted by cells. Using these probes, we can easily distinguish the differences in force-bearing integrins without perturbing adhesion biology and reveal that a strong force-bearing integrin cluster can serve as a 'mechanical pivot' to maintain focal adhesion architecture and facilitate its maturation. The benefits of the RSDTP include a high dynamic range, reversibility and single-molecule sensitivity, all of which will facilitate a better understanding of the molecular mechanisms of mechanobiology.
Collapse
Affiliation(s)
- Hongyun Li
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Chen Zhang
- College of Life Sciences, State Key Laboratory of Virology, Wuhan University, Wuhan, China
| | - Yuru Hu
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Pengxiang Liu
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Feng Sun
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Wei Chen
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Xinghua Zhang
- The Institute for Advanced Studies, Wuhan University, Wuhan, China.,College of Life Sciences, State Key Laboratory of Virology, Wuhan University, Wuhan, China
| | - Jie Ma
- School of Physics, Sun Yat-sen University, Guangzhou, China
| | - Wenxu Wang
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Liang Wang
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Piyu Wu
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Zheng Liu
- The Institute for Advanced Studies, Wuhan University, Wuhan, China.
| |
Collapse
|
43
|
Vasilaki D, Bakopoulou A, Tsouknidas A, Johnstone E, Michalakis K. Biophysical interactions between components of the tumor microenvironment promote metastasis. Biophys Rev 2021; 13:339-357. [PMID: 34168685 PMCID: PMC8214652 DOI: 10.1007/s12551-021-00811-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
During metastasis, tumor cells need to adapt to their dynamic microenvironment and modify their mechanical properties in response to both chemical and mechanical stimulation. Physical interactions occur between cancer cells and the surrounding matrix including cell movements and cell shape alterations through the process of mechanotransduction. The latter describes the translation of external mechanical cues into intracellular biochemical signaling. Reorganization of both the cytoskeleton and the extracellular matrix (ECM) plays a critical role in these spreading steps. Migrating tumor cells show increased motility in order to cross the tumor microenvironment, migrate through ECM and reach the bloodstream to the metastatic site. There are specific factors affecting these processes, as well as the survival of circulating tumor cells (CTC) in the blood flow until they finally invade the secondary tissue to form metastasis. This review aims to study the mechanisms of metastasis from a biomechanical perspective and investigate cell migration, with a focus on the alterations in the cytoskeleton through this journey and the effect of biologic fluids on metastasis. Understanding of the biophysical mechanisms that promote tumor metastasis may contribute successful therapeutic approaches in the fight against cancer.
Collapse
Affiliation(s)
- Dimitra Vasilaki
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Athina Bakopoulou
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Alexandros Tsouknidas
- Laboratory for Biomaterials and Computational Mechanics, Department of Mechanical Engineering, University of Western Macedonia, Kozani, Greece
| | | | - Konstantinos Michalakis
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
- Division of Graduate Prosthodontics, Tufts University School of Dental Medicine, Boston, MA USA
- University of Oxford, Oxford, UK
| |
Collapse
|
44
|
Kladko DV, Falchevskaya AS, Serov NS, Prilepskii AY. Nanomaterial Shape Influence on Cell Behavior. Int J Mol Sci 2021; 22:5266. [PMID: 34067696 PMCID: PMC8156540 DOI: 10.3390/ijms22105266] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 12/18/2022] Open
Abstract
Nanomaterials are proven to affect the biological activity of mammalian and microbial cells profoundly. Despite this fact, only surface chemistry, charge, and area are often linked to these phenomena. Moreover, most attention in this field is directed exclusively at nanomaterial cytotoxicity. At the same time, there is a large body of studies showing the influence of nanomaterials on cellular metabolism, proliferation, differentiation, reprogramming, gene transfer, and many other processes. Furthermore, it has been revealed that in all these cases, the shape of the nanomaterial plays a crucial role. In this paper, the mechanisms of nanomaterials shape control, approaches toward its synthesis, and the influence of nanomaterial shape on various biological activities of mammalian and microbial cells, such as proliferation, differentiation, and metabolism, as well as the prospects of this emerging field, are reviewed.
Collapse
Affiliation(s)
| | | | | | - Artur Y. Prilepskii
- International Institute “Solution Chemistry of Advanced Materials and Technologies”, ITMO University, 191002 Saint Petersburg, Russia; (D.V.K.); (A.S.F.); (N.S.S.)
| |
Collapse
|
45
|
Evolutionarily related small viral fusogens hijack distinct but modular actin nucleation pathways to drive cell-cell fusion. Proc Natl Acad Sci U S A 2021; 118:2007526118. [PMID: 33443166 DOI: 10.1073/pnas.2007526118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Fusion-associated small transmembrane (FAST) proteins are a diverse family of nonstructural viral proteins. Once expressed on the plasma membrane of infected cells, they drive fusion with neighboring cells, increasing viral spread and pathogenicity. Unlike viral fusogens with tall ectodomains that pull two membranes together through conformational changes, FAST proteins have short fusogenic ectodomains that cannot bridge the intermembrane gap between neighboring cells. One orthoreovirus FAST protein, p14, has been shown to hijack the actin cytoskeleton to drive cell-cell fusion, but the actin adaptor-binding motif identified in p14 is not found in any other FAST protein. Here, we report that an evolutionarily divergent FAST protein, p22 from aquareovirus, also hijacks the actin cytoskeleton but does so through different adaptor proteins, Intersectin-1 and Cdc42, that trigger N-WASP-mediated branched actin assembly. We show that despite using different pathways, the cytoplasmic tail of p22 can replace that of p14 to create a potent chimeric fusogen, suggesting they are modular and play similar functional roles. When we directly couple p22 with the parallel filament nucleator formin instead of the branched actin nucleation promoting factor N-WASP, its ability to drive fusion is maintained, suggesting that localized mechanical pressure on the plasma membrane coupled to a membrane-disruptive ectodomain is sufficient to drive cell-cell fusion. This work points to a common biophysical strategy used by FAST proteins to push rather than pull membranes together to drive fusion, one that may be harnessed by other short fusogens responsible for physiological cell-cell fusion.
Collapse
|
46
|
Haspinger DC, Klinge S, Holzapfel GA. Numerical analysis of the impact of cytoskeletal actin filament density alterations onto the diffusive vesicle-mediated cell transport. PLoS Comput Biol 2021; 17:e1008784. [PMID: 33939706 PMCID: PMC8130967 DOI: 10.1371/journal.pcbi.1008784] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 05/18/2021] [Accepted: 02/09/2021] [Indexed: 11/21/2022] Open
Abstract
The interior of a eukaryotic cell is a highly complex composite material which consists of water, structural scaffoldings, organelles, and various biomolecular solutes. All these components serve as obstacles that impede the motion of vesicles. Hence, it is hypothesized that any alteration of the cytoskeletal network may directly impact or even disrupt the vesicle transport. A disruption of the vesicle-mediated cell transport is thought to contribute to several severe diseases and disorders, such as diabetes, Parkinson’s and Alzheimer’s disease, emphasizing the clinical relevance. To address the outlined objective, a multiscale finite element model of the diffusive vesicle transport is proposed on the basis of the concept of homogenization, owed to the complexity of the cytoskeletal network. In order to study the microscopic effects of specific nanoscopic actin filament network alterations onto the vesicle transport, a parametrized three-dimensional geometrical model of the actin filament network was generated on the basis of experimentally observed filament densities and network geometries in an adenocarcinomic human alveolar basal epithelial cell. Numerical analyzes of the obtained effective diffusion properties within two-dimensional sampling domains of the whole cell model revealed that the computed homogenized diffusion coefficients can be predicted statistically accurate by a simple two-parameter power law as soon as the inaccessible area fraction, due to the obstacle geometries and the finite size of the vesicles, is known. This relationship, in turn, leads to a massive reduction in computation time and allows to study the impact of a variety of different cytoskeletal alterations onto the vesicle transport. Hence, the numerical simulations predicted a 35% increase in transport time due to a uniformly distributed four-fold increase of the total filament amount. On the other hand, a hypothetically reduced expression of filament cross-linking proteins led to sparser filament networks and, thus, a speed up of the vesicle transport. Many vital processes in our eukaryotic cells and organs require an astonishingly precise routing of intermediate products to various intra- and extracellular destinations using vesicles as transporters. This can be illustrated by numerous examples, such as the production and destruction of proteins, the export of neurotransmitters or insulin to the extracellular domain, etc. However, the inside of a cell is tightly packed with numerous structural scaffoldings (filaments), which serve as obstacles and impede the vesicle motion. It is thought that any disturbances of the vesicle-mediated cell transport contribute to numerous degenerative diseases and disorders, which highlights the clinical relevance for investigating this intracellular transport mechanism by developing computational models and performing experimental studies. In this study, we numerically quantified how different specific alterations of the filament density inside a human lung cell—due to changed mechanical loadings or genetic disorders of proteins being responsible for filament branching—affect the diffusion of vesicles inside the intracellular fluid. Therefore, based on the concept of homogenization, a computationally efficient numerical method was developed and utilized to simulate the diffusion of vesicles inside the whole cell, considering the detailed structural information of the filament network.
Collapse
Affiliation(s)
| | - Sandra Klinge
- Chair of Structural Mechanics and Analysis, TU Berlin, Berlin, Germany
| | - Gerhard A. Holzapfel
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
- Faculty of Engineering Science and Technology, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- * E-mail:
| |
Collapse
|
47
|
Cheah JS, Jacobs KA, Lai TW, Caballelo R, Yee JL, Ueda S, Heinrich V, Yamada S. Spatial proximity of proteins surrounding zyxin under force-bearing conditions. Mol Biol Cell 2021; 32:1221-1228. [PMID: 33909446 PMCID: PMC8351546 DOI: 10.1091/mbc.e19-10-0568] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Sensing physical forces is a critical first step in mechano-transduction of cells. Zyxin, a LIM domain-containing protein, is recruited to force-bearing actin filaments and is thought to repair and strengthen them. Yet, the precise force-induced protein interactions surrounding zyxin remain unclear. Using BioID analysis, we identified proximal proteins surrounding zyxin under normal and force-bearing conditions by label-free mass spectrometry analysis. Under force-bearing conditions, increased biotinylation of α-actinin 1, α-actinin 4, and AFAP1 were detected, and these proteins accumulated along force-bearing actin fibers independently from zyxin, albeit at a lower intensity than zyxin. VASP also accumulated along force-bearing actin fibers in a zyxin-dependent manner, but the biotinylation of VASP remained constant regardless of force, supporting the model of a free zyxin-VASP complex in the cytoplasm being corecruited to tensed actin fibers. In addition, ARHGAP42, a RhoA GAP, was also identified as a proximal protein of zyxin and colocalized with zyxin along contractile actin bundles. The overexpression of ARHGAP42 reduced the rate of small wound closure, a zyxin-dependent process. These results demonstrate that the application of proximal biotinylation can resolve the proximity and composition of protein complexes as a function of force, which had not been possible with traditional biochemical analysis.
Collapse
Affiliation(s)
- Joleen S Cheah
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616.,Biosciences Program, Stanford University, Stanford, CA 94305
| | - Kyle A Jacobs
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616
| | - Tzu Wei Lai
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616
| | - Reca Caballelo
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616
| | - Jacqueline L Yee
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616
| | - Shuji Ueda
- Graduate School of Agricultural Science, Kobe University, Kobe, Japan 657
| | - Volkmar Heinrich
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616
| | - Soichiro Yamada
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616
| |
Collapse
|
48
|
Li J, Jiang M, Su M, Tian L, Shi W, Yu C. Stretchable and Transparent Electrochemical Sensor Based on Nanostructured Au on Carbon Nanotube Networks for Real-Time Analysis of H 2O 2 Release from Cells. Anal Chem 2021; 93:6723-6730. [PMID: 33891403 DOI: 10.1021/acs.analchem.1c00336] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Various electrochemical biosensors have been developed for direct and real-time recording of biomolecules released from living cells. However, since these traditional electrodes are commonly rigid and nonflexible, in situ monitoring of biochemical signals while cell deformation occurs remains a great challenge. Herein, we report a facile approach for the development of a stretchable and transparent electrochemical cell-sensing platform based on Au nanostructures (nano-Au) and carbon nanotube (CNT) films embedded in PDMS (nano-Au/CNTs/PDMS). The sandwich-like nanostructured network of nano-Au/CNTs endows the sensor with excellent mechanical stability and electrochemical performance. The obtained nano-Au/CNTs/PDMS electrode displays desired performance for H2O2 detection with a wide linear range (20 nM-25.8 μM) and low detection limit (8 nM). Owing to good biocompatibility and flexibility, HeLa and human umbilical vein endothelial cells can be directly cultured on the electrode and real-time monitoring of H2O2 release from cells under their stretched state was realized. The proposed strategy demonstrated in this work provides an effective way for design of stretchable sensors and more opportunities for sensing biomolecules from mechanically sensitive cells.
Collapse
Affiliation(s)
- Jing Li
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| | - Mengyuan Jiang
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| | - Mengjie Su
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| | - Liang Tian
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| | - Weishan Shi
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| | - Chunmei Yu
- School of Public Health, Nantong University, Nantong 226019, P. R. China
| |
Collapse
|
49
|
Wang Y, Yang Y, Wang X, Yoshitomi T, Kawazoe N, Yang Y, Chen G. Micropattern-controlled chirality of focal adhesions regulates the cytoskeletal arrangement and gene transfection of mesenchymal stem cells. Biomaterials 2021; 271:120751. [PMID: 33740614 DOI: 10.1016/j.biomaterials.2021.120751] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023]
Abstract
Cell chirality has been demonstrated to be important for controlling cell functions. However, it is not clear how the chirality of the extracellular microenvironment regulates cell adhesion and cytoskeletal structures and therefore affects gene transfection. In this study, the chirality of focal adhesions and the cytoskeleton of single human mesenchymal stem cells (hMSCs) was controlled by specially designed micropatterns, and its influence on gene transfection was investigated. Micropatterns with different cell adhesion areas and swirling stripe lines were prepared by micropatterning fibronectin on polystyrene surfaces. The chiral micropatterns induced the formation of chiral focal adhesions and chiral cytoskeletal structures. Gene transfection efficiency was enhanced with increasing adhesion area, while hMSCs on left-handed and right-handed swirling micropatterns showed the same level of gene transfection. When the swirling angle was changed from 0°, 30°, and 60° to 90°, the gene transfection efficiency at a swirling angle of 60° was the lowest. The influence of cell chirality on gene transfection was strongly associated with cellular uptake capacity, DNA synthesis and cytoskeletal mechanics. The results demonstrated that cytoskeletal swirling had a significant influence on gene transfection.
Collapse
Affiliation(s)
- Yongtao Wang
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Yingjun Yang
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Xinlong Wang
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Toru Yoshitomi
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Naoki Kawazoe
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Yingnan Yang
- Graduate School of Life and Environmental Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Guoping Chen
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
50
|
Morales X, Cortés-Domínguez I, Ortiz-de-Solorzano C. Modeling the Mechanobiology of Cancer Cell Migration Using 3D Biomimetic Hydrogels. Gels 2021; 7:17. [PMID: 33673091 PMCID: PMC7930983 DOI: 10.3390/gels7010017] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/29/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
Understanding how cancer cells migrate, and how this migration is affected by the mechanical and chemical composition of the extracellular matrix (ECM) is critical to investigate and possibly interfere with the metastatic process, which is responsible for most cancer-related deaths. In this article we review the state of the art about the use of hydrogel-based three-dimensional (3D) scaffolds as artificial platforms to model the mechanobiology of cancer cell migration. We start by briefly reviewing the concept and composition of the extracellular matrix (ECM) and the materials commonly used to recreate the cancerous ECM. Then we summarize the most relevant knowledge about the mechanobiology of cancer cell migration that has been obtained using 3D hydrogel scaffolds, and relate those discoveries to what has been observed in the clinical management of solid tumors. Finally, we review some recent methodological developments, specifically the use of novel bioprinting techniques and microfluidics to create realistic hydrogel-based models of the cancer ECM, and some of their applications in the context of the study of cancer cell migration.
Collapse
Affiliation(s)
| | | | - Carlos Ortiz-de-Solorzano
- IDISNA, Ciberonc and Solid Tumors and Biomarkers Program, Center for Applied Medical Research, University of Navarra, 31008 Pamplona, Spain; (X.M.); (I.C.-D.)
| |
Collapse
|